1
|
Xavier LEMDS, Reis TCG, Martins ASDP, Santos JCDF, Bueno NB, Goulart MOF, Moura FA. Antioxidant Therapy in Inflammatory Bowel Diseases: How Far Have We Come and How Close Are We? Antioxidants (Basel) 2024; 13:1369. [PMID: 39594511 PMCID: PMC11590966 DOI: 10.3390/antiox13111369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Inflammatory bowel diseases (IBD) pose a growing public health challenge with unclear etiology and limited efficacy of traditional pharmacological treatments. Alternative therapies, particularly antioxidants, have gained scientific interest. This systematic review analyzed studies from MEDLINE, Cochrane, Web of Science, EMBASE, and Scopus using keywords like "Inflammatory Bowel Diseases" and "Antioxidants." Initially, 925 publications were identified, and after applying inclusion/exclusion criteria-covering studies from July 2015 to June 2024 using murine models or clinical trials in humans and evaluating natural or synthetic substances affecting oxidative stress markers-368 articles were included. This comprised 344 animal studies and 24 human studies. The most investigated antioxidants were polyphenols and active compounds from medicinal plants (n = 242; 70.3%). The review found a strong link between oxidative stress and inflammation in IBD, especially in studies on nuclear factor kappa B and nuclear factor erythroid 2-related factor 2 pathways. However, it remains unclear whether inflammation or oxidative stress occurs first in IBD. Lipid peroxidation was the most studied oxidative damage, followed by DNA damage. Protein damage was rarely investigated. The relationship between antioxidants and the gut microbiota was examined in 103 animal studies. Human studies evaluating oxidative stress markers were scarce, reflecting a major research gap in IBD treatment. PROSPERO registration: CDR42022335357 and CRD42022304540.
Collapse
Affiliation(s)
| | | | - Amylly Sanuelly da Paz Martins
- Postgraduate Studies at the Northeast Biotechnology Network (RENORBIO), Federal University of Alagoas (UFAL), Maceió 57072-970, AL, Brazil;
| | - Juliana Célia de Farias Santos
- Postgraduate Degree in Medical Sciences (PPGCM/UFAL), Federal University of Alagoas (UFAL), Maceió 57072-970, AL, Brazil;
| | - Nassib Bezerra Bueno
- Postgraduate Degree in Nutrition (PPGNUT), Federal University of Alagoas (UFAL), Maceió 57072-970, AL, Brazil; (L.E.M.d.S.X.); (N.B.B.)
| | - Marília Oliveira Fonseca Goulart
- Postgraduate Studies at the Northeast Biotechnology Network (RENORBIO), Federal University of Alagoas (UFAL), Maceió 57072-970, AL, Brazil;
- Institute of Chemistry and Biotechnology (IQB/UFAL), Federal University of Alagoas (UFAL), Maceió 57072-970, AL, Brazil
| | - Fabiana Andréa Moura
- Postgraduate Degree in Nutrition (PPGNUT), Federal University of Alagoas (UFAL), Maceió 57072-970, AL, Brazil; (L.E.M.d.S.X.); (N.B.B.)
- Postgraduate Degree in Medical Sciences (PPGCM/UFAL), Federal University of Alagoas (UFAL), Maceió 57072-970, AL, Brazil;
| |
Collapse
|
2
|
Lu Z, Liu J, Zhao L, Wang C, Shi F, Li Z, Liu X, Miao Z. Enhancement of oral bioavailability and anti-colitis effect of luteolin-loaded polymer micelles with RA (rosmarinic acid)-SS-mPEG as carrier. Drug Dev Ind Pharm 2023; 49:17-29. [PMID: 36730369 DOI: 10.1080/03639045.2023.2175850] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Polymer micelles were prepared (L-RSPMs) with luteolin and synthetic RA-SS-mPEG polymeric material before evaluation of their anti-inflammatory effect on 2, 4, 6-trinitro-benzene-sulfonic acid (TNBS)-induced ulcerative colitis (UC) model in rats. METHODS The synthetic RA-SS-mPEG was characterized with NMR spectroscopy, before preparation of luteolin-coated RA-SS-mPEG polymer micelles. The in vitro characterization and evaluation of the formulation were accomplished, couple with its pharmacokinetic parameters. The levels of PEG2, MDA, CRP and GSH, as well as concentrations of TNF-α, IL1-β, IL-6 and IL-10 in serum and colon tissue were detected via ELISA kit. The degree of colon injury and inflammation was evaluated via histopathologic examination. RESULTS L-RSPMs displayed small average droplet size (133.40 ± 4.52 nm), uniformly dispersed (PDI: 0.163 ± 0.011), good stability, slow release and enhanced solubility. We observed 353.28% increase in the relative bioavailability of L-RSPMs compared to free luteolin, while the half-life of the micelle was extended by 6.16h. Compared to model (M) group, luteolin (low and high doses) and L-RSPMs (low and high doses) significantly reduced levels of MDA, PEG2, CRP, TNF-α, IL-6 and IL-1β in colon tissue and serum of colitic rats but dose dependently increased IL-10 and SOD levels (p < 0.01). Histopathologic examination of colon showed that luteolin (low and high doses) and L-RSPMs (low and high doses) improved colonic inflammation in colitic rats to varying degrees compared to M group. CONCLUSION L-RSPMs could improve TNBS-induced colon inflammation by enhancing bioavailability, promoting antioxidant effects and regulating cytokine release, which may become a potential agent for UC treatment in clinical settings.
Collapse
Affiliation(s)
- Zhaomin Lu
- Department of Gastroenterology, The Second People's Hospital of Zhangjiagang, Zhangjiagang, China
| | - Juan Liu
- Department of Gastroenterology, The Second People's Hospital of Zhangjiagang, Zhangjiagang, China
| | - Liangjian Zhao
- Department of Gastroenterology, The Second People's Hospital of Zhangjiagang, Zhangjiagang, China
| | - Chenli Wang
- Department of Gastroenterology, The Second People's Hospital of Zhangjiagang, Zhangjiagang, China
| | - Feng Shi
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Zhengqi Li
- Department of Gastroenterology, The Second People's Hospital of Zhangjiagang, Zhangjiagang, China
| | - Xuesong Liu
- Department of Gastroenterology, The Second People's Hospital of Zhangjiagang, Zhangjiagang, China
| | - Zhiwei Miao
- Department of Gastroenterology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China
| |
Collapse
|
3
|
Wilhelm EA, Soares PS, Reis AS, Motta KP, Lemos BB, Domingues WB, Blödorn EB, Araujo DR, Barcellos AM, Perin G, Soares MP, Campos VF, Luchese C. Se-[(2,2-Dimethyl-1,3-dioxolan-4-yl)methyl] 4-Chlorobenzoselenolate Attenuates Inflammatory Response, Nociception, and Affective Disorders Related to Rheumatoid Arthritis in Mice. ACS Chem Neurosci 2021; 12:3760-3771. [PMID: 34553902 DOI: 10.1021/acschemneuro.1c00512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Despite major advances, not all patients achieve rheumatoid arthritis (RA) remission, thus highlighting a pressing need for new therapeutic treatments. Given this scenario, this study sought to evaluate Se-[(2,2-dimethyl-1,3-dioxolan-4-yl)methyl] 4-chlorobenzoselenolate (Se-DMC) potential on a complete Freund's adjuvant (CFA)-induced unilateral arthritis model. The effects of Se-DMC (5 mg/kg; oral dose) and meloxicam (5 mg/kg; oral dose), both administered to animals daily for 14 days, on paw edema, mechanical sensitivity, neurobehavioral deficits (anxiogenic- and depressive-like behaviors), Na+/K+-ATPase activity, oxidative stress, and inflammation were evaluated in male Swiss mice exposed to CFA (intraplantar injection of 0.1 mL; 10 mg/mL). Se-DMC reduced the paw withdrawal threshold and CFA-induced paw edema. Histopathological results revealed the antiedematogenic potential of the compound, which was evidenced by lower quantities of dilated lymphatic vessels compared with the CFA group. Se-DMC reduced mRNA relative expression levels of tumor necrosis factor-α (TNF-α) and nuclear factor-κB (NF-κB) in the hippocampus and paw of CFA mice. The CFA-induced anxiogenic- and depressive-like behaviors were reversed by Se-DMC to the control levels in the elevated plus-maze and tail suspension tests. Se-DMC reduced the paw reactive species levels and restored the superoxide dismutase (hippocampus and paw) and Na+/K+-ATPase (hippocampus) activities previously increased by CFA. Moreover, CFA administration inhibited serum creatinine kinase activity, albeit the Se-DMC effects did not appear to involve the modulation of this enzyme and were equal to or greater than meloxicam. Se-DMC attenuates CFA-induced inflammatory response, nociception, and neurobehavioral deficits in mice.
Collapse
Affiliation(s)
- Ethel A. Wilhelm
- Laboratório de Pesquisa em Farmacologia Bioquímica, CCQFA, Universidade Federal de Pelotas, UFPel, Pelotas 96010-900, Brazil
| | - Paola S. Soares
- Laboratório de Pesquisa em Farmacologia Bioquímica, CCQFA, Universidade Federal de Pelotas, UFPel, Pelotas 96010-900, Brazil
| | - Angélica S. Reis
- Laboratório de Pesquisa em Farmacologia Bioquímica, CCQFA, Universidade Federal de Pelotas, UFPel, Pelotas 96010-900, Brazil
| | - Ketlyn P. Motta
- Laboratório de Pesquisa em Farmacologia Bioquímica, CCQFA, Universidade Federal de Pelotas, UFPel, Pelotas 96010-900, Brazil
| | - Briana B. Lemos
- Laboratório de Pesquisa em Farmacologia Bioquímica, CCQFA, Universidade Federal de Pelotas, UFPel, Pelotas 96010-900, Brazil
| | - William B. Domingues
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Laboratório de Genômica Estrutural, Biotecnologia, Universidade Federal de Pelotas, UFPel, Campus Capão do Leão, Pelotas 96010-900, RS, Brazil
| | - Eduardo B. Blödorn
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Laboratório de Genômica Estrutural, Biotecnologia, Universidade Federal de Pelotas, UFPel, Campus Capão do Leão, Pelotas 96010-900, RS, Brazil
| | - Daniela R. Araujo
- Laboratório de Síntese Orgânica Limpa, CCQFA, Universidade Federal de Pelotas—UFPel, Pelotas 96010-900, Brazil
| | - Angelita M. Barcellos
- Laboratório de Síntese Orgânica Limpa, CCQFA, Universidade Federal de Pelotas—UFPel, Pelotas 96010-900, Brazil
| | - Gelson Perin
- Laboratório de Síntese Orgânica Limpa, CCQFA, Universidade Federal de Pelotas—UFPel, Pelotas 96010-900, Brazil
| | - Mauro P. Soares
- Laboratório Regional de Diagnóstico, Faculdade de Veterinária, Universidade Federal de Pelotas, UFPel, Pelotas 96010-900, Brazil
| | - Vinicius F. Campos
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Laboratório de Genômica Estrutural, Biotecnologia, Universidade Federal de Pelotas, UFPel, Campus Capão do Leão, Pelotas 96010-900, RS, Brazil
| | - Cristiane Luchese
- Laboratório de Pesquisa em Farmacologia Bioquímica, CCQFA, Universidade Federal de Pelotas, UFPel, Pelotas 96010-900, Brazil
| |
Collapse
|
4
|
da Cunha VP, Preisser TM, Santana MP, Machado DCC, Pereira VB, Miyoshi A. Invasive Lactococcus lactis producing mycobacterial Hsp65 ameliorates intestinal inflammation in acute TNBS-induced colitis in mice by increasing the levels of the cytokine IL-10 and secretory IgA. J Appl Microbiol 2020; 129:1389-1401. [PMID: 32473073 DOI: 10.1111/jam.14695] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/22/2020] [Accepted: 04/30/2020] [Indexed: 12/22/2022]
Abstract
AIMS To investigate the anti-inflammatory activity of an invasive and Hp65-producing strain Lactococcus lactis NCDO2118 FnBPA+ (pXYCYT:Hsp65) in acute 2,4,6-trinitrobenzene sulphonic acid (TNBS)-induced colitis in mice as an innovative therapeutic strategy against Crohn's disease (CD). METHODS AND RESULTS The pXYCYT:Hsp65 plasmid was transformed into the L. lactis NCDO2118 FnBPA+ strain, resulting in the L. lactis NCDO2118 FnBPA+ (pXYCYT:Hsp65) strain. Then, the functionality of the strain was evaluated in vitro for Hsp65 production by Western blotting and for invasion into Caco-2 cells. The results demonstrated that the strain was able to produce Hsp65 and efficiently invade eukaryotic cells. Subsequently, in vivo, the anti-inflammatory capacity of the recombinant strain was evaluated in colitis induced with TNBS in BALB/c mice. Oral administration of the recombinant strain was able to attenuated the severity of colitis by mainly reducing IL-12 and IL-17 levels and increasing IL-10 and secretory immunoglobulin A levels. CONCLUSIONS The L. lactis NCDO2118 FnBPA+ (pXYCYT:Hsp65) strain contributed to a reduction in inflammatory damage in experimental CD. SIGNIFICANCE AND IMPACT OF THE STUDY This study, which used L. lactis for the production and delivery of Hsp65, has scientific relevance because it shows the efficacy of this new strategy based on therapeutic protein delivery into mammalian enterocytes.
Collapse
Affiliation(s)
- V P da Cunha
- Laboratory of Genetic Technology, Department of Ecology, Genetics and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - T M Preisser
- Laboratory of Genetic Technology, Department of Ecology, Genetics and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - M P Santana
- Laboratory of Genetic Technology, Department of Ecology, Genetics and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - D C C Machado
- Center for Gastrointestinal Biology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - V B Pereira
- Laboratory of Genetic Technology, Department of Ecology, Genetics and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - A Miyoshi
- Laboratory of Genetic Technology, Department of Ecology, Genetics and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
5
|
Silva I, Pinto R, Mateus V. Preclinical Study in Vivo for New Pharmacological Approaches in Inflammatory Bowel Disease: A Systematic Review of Chronic Model of TNBS-Induced Colitis. J Clin Med 2019; 8:jcm8101574. [PMID: 31581545 PMCID: PMC6832474 DOI: 10.3390/jcm8101574] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 09/19/2019] [Accepted: 09/25/2019] [Indexed: 12/14/2022] Open
Abstract
The preclinical studies in vivo provide means of characterizing physiologic interactions when our understanding of such processes is insufficient to allow replacement with in vitro systems and play a pivotal role in the development of a novel therapeutic drug cure. Chemically induced colitis models are relatively easy and rapid to develop. The 2,4,6-trinitrobenzenesulfonic acid (TNBS) colitis model is one of the main models in the experimental studies of inflammatory bowel disease (IBD) since inflammation induced by TNBS mimics several features of Crohn’s disease. This review aims to summarize the existing literature and discuss different protocols for the induction of chronic model of TNBS-induced colitis. We searched MEDLINE via Pubmed platform for studies published through December 2018, using MeSH terms (Crohn Disease.kw) OR (Inflammatory Bowel Diseases.kw) OR (Colitis, Ulcerative.kw) AND (trinitrobenzenesulfonic acid.kw) AND (disease models, animal.kw) AND (mice.all). The inclusion criteria were original articles, preclinical studies in vivo using mice, chronic model of colitis, and TNBS as the inducer of colitis and articles published in English. Chronic TNBS-induced colitis is made with multiple TNBS intrarectal administrations in an average dose of 1.2 mg using a volume lower than 150 μL in 50% ethanol. The strains mostly used are Balb/c and C57BL/6 with 5–6 weeks. To characterize the preclinical model the parameters more used include body weight, stool consistency and morbidity, inflammatory biomarkers like interferon (IFN)-γ, myeloperoxidase (MPO), tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-10, presence of ulcers, thickness or hyperemia in the colon, and histological evaluation of the inflammation. Experimental chronic colitis is induced by multiple rectal instillations of TNBS increasing doses in ethanol using Balb/c and C57BL/6 mice.
Collapse
Affiliation(s)
- Inês Silva
- H&TRC–Health and Technology Research Center, ESTeSL–Lisbon School of Health Technology, Instituto Politécnico de Lisboa, 1990-096 Lisbon, Portugal;
- iMed.ULisboa, Faculdade de Farmácia, Universidade de Lisboa, 1990-096 Lisboa, Portugal;
| | - Rui Pinto
- iMed.ULisboa, Faculdade de Farmácia, Universidade de Lisboa, 1990-096 Lisboa, Portugal;
- JCS, Dr. Joaquim Chaves, Laboratório de Análises Clínicas, Miraflores, 1495-069 Algés, Portugal
| | - Vanessa Mateus
- H&TRC–Health and Technology Research Center, ESTeSL–Lisbon School of Health Technology, Instituto Politécnico de Lisboa, 1990-096 Lisbon, Portugal;
- iMed.ULisboa, Faculdade de Farmácia, Universidade de Lisboa, 1990-096 Lisboa, Portugal;
- Correspondence: ; Tel.: +351-218-980-400; Fax: +351-218-980-460
| |
Collapse
|
6
|
Dou YX, Zhou JT, Wang TT, Huang YF, Chen VP, Xie YL, Lin ZX, Gao JS, Su ZR, Zeng HF. Self-nanoemulsifying drug delivery system of bruceine D: a new approach for anti-ulcerative colitis. Int J Nanomedicine 2018; 13:5887-5907. [PMID: 30319255 PMCID: PMC6167998 DOI: 10.2147/ijn.s174146] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background Bruceine D (BD) is a major bioactive component isolated from the traditional Chinese medicinal plant Brucea javanica which has been widely utilized to treat dysentery (also known as ulcerative colitis [UC]). Methods To improve the water solubility and absolute bioavailability of BD, we developed a self-nanoemulsifying drug delivery system (SNEDDS) composing of MCT (oil), Solutol HS-15 (surfactant), propylene glycol (co-surfactant) and BD. The physicochemical properties and pharmacokinetics of BD-SNEDDS were characterized, and its anti-UC activity and potential mechanism were evaluated in TNBS-induced UC rat model. Results The prepared nanoemulsion has multiple beneficial aspects including small mean droplet size, low polydispersity index (PDI), high zeta potential (ZP) and excellent stability. Transmission electron microscopy showed that nanoemulsion droplets contained uniform shape and size of globules. Pharmacokinetic studies demonstrated that BD-SNEDDS exhibited enhanced pharmacokinetic parameters as compared with BD-suspension. Moreover, BD-SNEDDS significantly restored the colon length and body weight, reduced disease activity index (DAI) and colon pathology, decreased histological scores, diminished oxidative stress, and suppressed TLR4, MyD88, TRAF6, NF-κB p65 protein expressions in TNBS-induced UC rat model. Conclusion These results demonstrated that BD-SNEDDS exhibited highly improved oral bioavailability and advanced anti-UC efficacy. In conclusion, our current results provided a foundation for further research of BD-SNEDDS as a potential complementary therapeutic agent for UC treatment.
Collapse
Affiliation(s)
- Yao-Xing Dou
- Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China.,Department of Pharmacy, The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China,
| | - Jiang-Tao Zhou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China.,Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Tong-Tong Wang
- Department of Pharmacy, The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China,
| | - Yan-Feng Huang
- Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| | - Vicky Ping Chen
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - You-Liang Xie
- Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| | - Zhi-Xiu Lin
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Jian-Sheng Gao
- Guangzhou Baiyunshan Mingxing Pharmaceutical Co. Ltd., Guangzhou, People's Republic of China
| | - Zi-Ren Su
- Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| | - Hui-Fang Zeng
- Department of Pharmacy, The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China,
| |
Collapse
|
7
|
Munchen TS, Sonego MS, de Souza D, Dornelles L, Seixas FK, Collares T, Piccoli BC, da Silva FD, da Rocha JBT, Quoos N, Rodrigues OED. New 3’‐Triazolyl‐5’‐aryl‐chalcogenothymidine: Synthesis and Anti‐oxidant and Antiproliferative Bladder Carcinoma (5637) Activity. ChemistrySelect 2018. [DOI: 10.1002/slct.201800156] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Taiana S. Munchen
- LabSelen-NanoBio – Departamento de QuímicaUniversidade Federal de Santa Maria RS - CEP 97105–900 – Brazil
| | - Mariana S. Sonego
- Programa de Pós-Graduação em Biotecnologia (PPGB)Biotecnologia/Centro de Desenvolvimento TecnológicoGrupo de Pesquisa em Oncologia Celular e Molecular (GPO)Laboratório de Biotecnologia do Câncer Universidade Federal de PelotasCampus Universitário s/n Capão do Leão-RS Brasil Cep: 96010–900
| | - Diego de Souza
- LabSelen-NanoBio – Departamento de QuímicaUniversidade Federal de Santa Maria RS - CEP 97105–900 – Brazil
| | - Luciano Dornelles
- LabSelen-NanoBio – Departamento de QuímicaUniversidade Federal de Santa Maria RS - CEP 97105–900 – Brazil
| | - Fabiana K. Seixas
- Programa de Pós-Graduação em Biotecnologia (PPGB)Biotecnologia/Centro de Desenvolvimento TecnológicoGrupo de Pesquisa em Oncologia Celular e Molecular (GPO)Laboratório de Biotecnologia do Câncer Universidade Federal de PelotasCampus Universitário s/n Capão do Leão-RS Brasil Cep: 96010–900
| | - Tiago Collares
- Programa de Pós-Graduação em Biotecnologia (PPGB)Biotecnologia/Centro de Desenvolvimento TecnológicoGrupo de Pesquisa em Oncologia Celular e Molecular (GPO)Laboratório de Biotecnologia do Câncer Universidade Federal de PelotasCampus Universitário s/n Capão do Leão-RS Brasil Cep: 96010–900
| | - Bruna C. Piccoli
- Departamento de Bioquímica e Biologia MolecularCentro de Ciências Naturais e ExatasUniversidade Federal de Santa Maria Santa Maria CEP 97105–900 Brazil
| | - Fernanda D'Ávila da Silva
- Departamento de Bioquímica e Biologia MolecularCentro de Ciências Naturais e ExatasUniversidade Federal de Santa Maria Santa Maria CEP 97105–900 Brazil
| | - João Batista T. da Rocha
- Departamento de Bioquímica e Biologia MolecularCentro de Ciências Naturais e ExatasUniversidade Federal de Santa Maria Santa Maria CEP 97105–900 Brazil
| | - Natália Quoos
- LabSelen-NanoBio – Departamento de QuímicaUniversidade Federal de Santa Maria RS - CEP 97105–900 – Brazil
| | - Oscar E. D. Rodrigues
- LabSelen-NanoBio – Departamento de QuímicaUniversidade Federal de Santa Maria RS - CEP 97105–900 – Brazil
| |
Collapse
|
8
|
Morinda citrifolia (Noni) Fruit Juice Reduces Inflammatory Cytokines Expression and Contributes to the Maintenance of Intestinal Mucosal Integrity in DSS Experimental Colitis. Mediators Inflamm 2017; 2017:6567432. [PMID: 28194046 PMCID: PMC5282445 DOI: 10.1155/2017/6567432] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 12/09/2016] [Accepted: 12/25/2016] [Indexed: 12/12/2022] Open
Abstract
Morinda citrifolia L. (noni) has been shown to treat different disorders. However, data concerning its role in the treatment of intestinal inflammation still require clarification. In the current study, we investigated the effects of noni fruit juice (NFJ) in the treatment of C57BL/6 mice, which were continuously exposed to dextran sulfate sodium (DSS) for 9 consecutive days. NFJ consumption had no impact on the reduction of the clinical signs of the disease or on weight loss. Nonetheless, when a dilution of 1 : 10 was used, the intestinal architecture of the mice was preserved, accompanied by a reduction in the inflammatory infiltrate. Regardless of the concentration of NFJ, a decrease in both the activity of myeloperoxidase and the key inflammatory cytokines, TNF-α and IFN-γ, was also observed in the intestine. Furthermore, when NFJ was diluted 1 : 10 and 1 : 100, a reduction in the production of nitric oxide and IL-17 was detected in gut homogenates. Overall, the treatment with NFJ was effective in different aspects associated with disease progression and worsening. These results may point to noni fruit as an important source of anti-inflammatory molecules with a great potential to inhibit the progression of inflammatory diseases, such as inflammatory bowel disease.
Collapse
|
9
|
Hong HS, Hwang DY, Park JH, Kim S, Seo EJ, Son Y. Substance-P alleviates dextran sulfate sodium-induced intestinal damage by suppressing inflammation through enrichment of M2 macrophages and regulatory T cells. Cytokine 2016; 90:21-30. [PMID: 27750083 DOI: 10.1016/j.cyto.2016.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 10/04/2016] [Accepted: 10/06/2016] [Indexed: 12/12/2022]
Abstract
Intestinal inflammation alters immune responses in the mucosa and destroys colon architecture, leading to serious diseases such as inflammatory bowel disease (IBD). Thus, regulation of inflammation is regarded as the ultimate therapy for intestinal disease. Substance-P (SP) is known to mediate proliferation, migration, and cellular senescence in a variety of cells. SP was found to mobilize stem cells from bone marrow to the site of injury and to suppress inflammatory responses by inducing regulatory T cells (Tregs) and M2 macrophages. In this study, we explored the effects of SP in a dextran sodium sulfate (DSS)-induced intestine damage model. The effects of SP were evaluated by analyzing crypt structures, proliferating cells within the colon, cytokine secretion profiles, and immune cells population in the spleen/mesenteric lymph nodes in vivo. DSS treatment provoked an inflammatory response with loss of crypts in the intestines of experimental mice. This response was associated with high levels of inflammatory cytokines such as TNF-α and IL-17, and low levels of Tregs and M2 macrophages, leading to severely damaged tissue structure. However, SP treatment inhibited inflammatory responses by modulating cytokine production as well as the balance of Tregs/Th 17 cells and the M1/M2 transition in lymphoid organs, leading to accelerated tissue repair. Collectively, our data indicate that SP can promote the regeneration of tissue following damage by DSS treatment, possibly by modulating immune response. Our results propose SP as a candidate therapeutic for intestine-related inflammatory diseases.
Collapse
Affiliation(s)
- Hyun Sook Hong
- College of Medicine/East-West Medical Research Institute, Kyung Hee University, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea.
| | - Dae Yeon Hwang
- Department of Medicine, Graduate School, Kyung Hee University, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Ju Hyeong Park
- Graduate School of Biotechnology & Department of Genetic Engineering, College of Life Science, Kyung Hee University, Seochun-dong, Kiheung-ku, Yong In 441-706, Republic of Korea
| | - Suna Kim
- Graduate School of Biotechnology & Department of Genetic Engineering, College of Life Science, Kyung Hee University, Seochun-dong, Kiheung-ku, Yong In 441-706, Republic of Korea
| | - Eun Jung Seo
- Department of Medicine, Graduate School, Kyung Hee University, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Youngsook Son
- Graduate School of Biotechnology & Department of Genetic Engineering, College of Life Science, Kyung Hee University, Seochun-dong, Kiheung-ku, Yong In 441-706, Republic of Korea.
| |
Collapse
|