Volume 13 Issue 12
Dec.  2023
Turn off MathJax
Article Contents
Gopalsamy Rajiv Gandhi, Thiruchenduran Mohana, Kumaraswamy Athesh, Varghese Edwin Hillary, Alan Bruno Silva Vasconcelos, Mariana Nobre Farias de Franca, Monalisa Martins Montalvão, Stanislaus Antony Ceasar, Gnanasekaran Jothi, Gurunagarajan Sridharan, Ricardo Queiroz Gurgel, Baojun Xu. Anti-inflammatory natural products modulate interleukins and their related signaling markers in inflammatory bowel disease: A systematic review[J]. Journal of Pharmaceutical Analysis, 2023, 13(12): 1408-1428. doi: 10.1016/j.jpha.2023.09.012
Citation: Gopalsamy Rajiv Gandhi, Thiruchenduran Mohana, Kumaraswamy Athesh, Varghese Edwin Hillary, Alan Bruno Silva Vasconcelos, Mariana Nobre Farias de Franca, Monalisa Martins Montalvão, Stanislaus Antony Ceasar, Gnanasekaran Jothi, Gurunagarajan Sridharan, Ricardo Queiroz Gurgel, Baojun Xu. Anti-inflammatory natural products modulate interleukins and their related signaling markers in inflammatory bowel disease: A systematic review[J]. Journal of Pharmaceutical Analysis, 2023, 13(12): 1408-1428. doi: 10.1016/j.jpha.2023.09.012

Anti-inflammatory natural products modulate interleukins and their related signaling markers in inflammatory bowel disease: A systematic review

doi: 10.1016/j.jpha.2023.09.012
Funds:

The authors would like to thank Rajagiri College of Social Sciences (Autonomous), Kochi, India for providing extensive research support. This study received funding from Rajagiri College of Social Sciences in the form of seed funding for executing Major Research Projects (Project No.: RCSS/IQAC/BB-S33/2023/133). Two United International College (UIC) research grants provided by BNU-HKBU United International College, Zhuhai, China jointly support this work (Grant Nos.: UICR0200007-23 and UICR202107).

  • Received Date: Apr. 20, 2023
  • Accepted Date: Sep. 18, 2023
  • Rev Recd Date: Sep. 17, 2023
  • Publish Date: Sep. 22, 2023
  • This review aims to identify in vivo studies investigating the potential of plant substances and their natural molecules in managing inflammatory bowel disease (IBD). Specifically, the objective is to examine the impact of these substances on interleukins and other key inflammatory signaling markers. Relevant articles published up to December 2022 were identified through a search of the PubMed, Scopus, Web of Science, and Embase databases. The search used keywords including “inflammatory bowel disease”, “medicinal plants”, “natural molecules”, “anti-inflammatory”, and “ulcerative colitis”, and identified 1,878 potentially relevant articles, of which 89 were included in this review after completion of the selection process. This study provides preclinical data on natural products (NPs) that can potentially treat IBD, including ulcerative colitis. The main actions of these NPs relate to their effects on nuclear factor kappa B (NF-κB), the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling pathway, the regulation of T helper 17/regulatory T cells balance, and oxidative stress. The ability of these NPs to inhibit intestinal inflammation appears to be dependent on lowering levels of the pro-inflammatory cytokines tumor necrosis factor-alpha (TNF-α), interleukin (IL)-1β, and IL-17, via the Jun N-terminal kinase (JNK)1, NF-κβ-p65, and STAT3 pathways. In addition, NPs were shown to reduce oxidative stress and the severity of ulcerative colitis, as well as increase the activity of antioxidant enzymes. These actions suggest that NPs represent a promising treatment for IBD, and potentially have greater efficacy and safety than current treatments.
  • loading
  • [1]
    S.M.A. Bastaki, N. Amir, E. Adeghate, et al., Nerolidol, a sesquiterpene, attenuates oxidative stress and inflammation in acetic acid-induced colitis in rats, Mol. Cell. Biochem. 476 (2021) 3497-3512.
    [2]
    N. Eltantawy, I.A.E. El-Zayyadi, A.A. Elberry, et al., A review article of infammatory bowel disease treatment and pharmacogenomics, Beni-Suef Univ. J. Basic Appl. Sci. 12 (2023), 35.
    [3]
    G.G. Kaplan, J.W. Windsor, The four epidemiological stages in the global evolution of inflammatory bowel disease, Nat. Rev. Gastroenterol. Hepatol. 18 (2021) 56-66.
    [4]
    X. Qin, C. Pan, Q. Cai, et al., Assessing the effect of interaction between gut microbiome and inflammatory bowel disease on the risks of depression, Brain Behav. Immun. Health 26 (2022), 100557.
    [5]
    T. Raine, S. Bonovas, J. Burisch, et al., ECCO guidelines on therapeutics in ulcerative colitis: medical treatment, J. Crohns Colitis 16 (2022) 2-17.
    [6]
    M. Olfatifar, M.R. Zali, M.A. Pourhoseingholi, et al., The emerging epidemic of inflammatory bowel disease in Asia and Iran by 2035: a modeling study, BMC Gastroenterol. 21 (2021), 204.
    [7]
    V.Q. Nguyen, K. Eden, H.A. Morrison, et al., Noncanonical NF-κB signaling upregulation in inflammatory bowel disease patients is associated with loss of response to anti-TNF agents, Front. Pharmacol. 12 (2021), 655887.
    [8]
    A. Saez, B. Herrero-Fernandez, R. Gomez-Bris, et al., Pathophysiology of inflammatory bowel disease: innate immune system, Int. J. Mol. Sci. 24 (2023), 1526.
    [9]
    P. Lu, Y. Zhao, Targeting NF-κB pathway for treating ulcerative colitis: comprehensive regulatory characteristics of Chinese medicines, Chin. Med. 15 (2020), 15.
    [10]
    L.F. Laurindo, A.R.O. dos Santos, A.C.A. de Carvalho, Phytochemicals and regulation of NF-κB in inflammatory bowel diseases: an overview of in vitro and in vivo effects, Metabolites 13 (2023), 96.
    [11]
    F. Cordes, D. Foell, J.N. Ding, et al., Differential regulation of JAK/STAT-signaling in patients with ulcerative colitis and Crohn's disease, World J. Gastroenterol. 26 (2020) 4055-4075.
    [12]
    L. Duan, S. Cheng, L. Li, et al., Natural anti-inflammatory compounds as drug candidates for inflammatory bowel disease, Front. Pharmacol. 12 (2021), 684486.
    [13]
    X. Hu, J. li, M. Fu, et al., The JAK/STAT signaling pathway: from bench to clinic, Signal Transduct. Target Ther. 6 (2021), 402.
    [14]
    Y. Zhou, D. Wang, W. Yan, Treatment effects of natural products on inflammatory bowel disease in vivo and their mechanisms: based on animal experiments, Nutrients 15 (2023), 1031.
    [15]
    S.Y. Moon, K.D. Kim, J. Yoo, et al., Phytochemicals targeting JAK-STAT pathways in inflammatory bowel disease: insights from animal models, Molecules 26 (2021), 2824.
    [16]
    J. Yan, M. Luo, Z. Chen, et al., The function and role of the Th17/Treg cell balance in inflammatory bowel disease, J. Immunol. Res. 2020 (2020), 8813558.
    [17]
    L. Rocamora-Reverte, F.L. Melzer, R. Wurzner, et al., The complex role of regulatory T cells in immunity and aging, Front. Immunol. 11 (2021), 616949.
    [18]
    X. Chen, J.J. Oppenheim, Th17 cells and Tregs: unlikely allies, J. Leukoc. Biol. 95 (2014) 723-731.
    [19]
    Y. Chen, W. Cui, X. Li, et al., Interaction between commensal bacteria, immune response and the intestinal barrier in inflammatory bowel disease, Front. Immunol. 12 (2021), 761981.
    [20]
    J. Yan, M. Luo, Z. Chen, et al., The function and role of the Th17/treg cell balance in inflammatory bowel disease, J. Immunol. Res. 2020 (2020) 1-8.
    [21]
    Y. Lin, X. Zheng, J. Chen, et al., Protective effect of Bruguiera gymnorrhiza (L.) lam. fruit on dextran sulfate sodium-induced ulcerative colitis in mice: role of Keap1/Nrf2 pathway and gut microbiota, Front. Pharmacol. 10 (2020), 1602.
    [22]
    R. Carreras Torres, G. Ibanez Sanz, M. Obon Santacana, et al., Identifying environmental risk factors for infammatory bowel diseases: a Mendelian randomization study, Sci. Rep. 10 (2020), 19273.
    [23]
    R. Rajbhandari, S. Blakemore, N. Gupta, Crohn's disease among the poorest billion: burden of Crohn's disease in low and lower middle income countries, Dig. Dis. Sci. 68 (2023) 1226-1236.
    [24]
    M. Gupta, V. Mishra, M. Gulati, et al., Natural compounds as safe therapeutic options for ulcerative colitis, Infammopharmacology 30 (2022) 397-434.
    [25]
    L.B. Sina, C.A. Secco, M. Blazevic, et al., Hybrid forecasting methods - a systematic review, Electronics 12 (2023), 2019.
    [26]
    D. Moher, A. Liberati, J. Tetzlaff, et al., Preferred reporting items for systematic reviews and meta-analyses: the PRISMA Statement, Open Med. 3 (2009) e123-e130.
    [27]
    J.O. Oladele, J.C. Anyim, O.M. Oyeleke, et al., Telfairia occidentalis mitigates dextran sodium sulfate-induced ulcerative colitis in rats via suppression of oxidative stress, lipid peroxidation, and inflammation, J. Food Biochem. 45 (2021), e13873.
    [28]
    C. Li, N. Dong, B. Wu, et al., Dihydroberberine, an isoquinoline alkaloid, exhibits protective effect against dextran sulfate sodium-induced ulcerative colitis in mice, Phytomedicine 90 (2021), 153631.
    [29]
    L. Lian, S. Zhang, Z. Yu, et al., The dietary freeze-dried fruit powder of Actinidia arguta ameliorates dextran sulphate sodium-induced ulcerative colitis in mice by inhibiting the activation of MAPKs, Food Funct. 10 (2019) 5768-5778.
    [30]
    A. Atta, S. Mouneir, S. Nasr, et al., Phytochemical studies and anti-ulcerative colitis effect of Moringa oleifera seeds and Egyptian propolis methanol extracts in a rat model, Asian Pac. J. Trop. Biomed. 9 (2019), 98.
    [31]
    A. Riaz, R.A. Khan, S. Afroz, et al., Prophylactic and therapeutic effect of Punica granatum in trinitrobenzene sulfonic acid induced inflammation in rats, Pak. J. Pharm. Sci. 30 (2017) 155-162.
    [32]
    S. Guo, W. Geng, S. Chen, et al., Ginger alleviates DSS-induced ulcerative colitis severity by improving the diversity and function of gut microbiota, Front. Pharmacol. 12 (2021), 632569.
    [33]
    D.D. Park, H.W. Yum, X. Zhong, et al., Perilla frutescens extracts protects against dextran sulfate sodium-induced murine colitis: NF-κB, STAT3, and Nrf2 as putative targets, Front. Pharmacol. 8 (2017), 482.
    [34]
    A. Sido, S. Radhakrishnan, S.W. Kim, et al., A food-based approach that targets interleukin-6, a key regulator of chronic intestinal inflammation and colon carcinogenesis, J. Nutr. Biochem. 43 (2017) 11-17.
    [35]
    S. Subramanya, G. Chaudhary, U. Mahajan, et al., Protective effect of Lagerstroemia speciosa against dextran sulfate sodium induced ulcerative colitis in C57BL/6 mice 65th International Congress and Annual Meeting of the Society for medicinal plant and natural product research (GA 2017)", "Planta Medica International Open. September 3-7, 2017. Basel, Switzerland. Georg Thieme Verlag KG, (2017).
    [36]
    X. Wu, L. Xue, A. Tata, et al., Bioactive components of polyphenol-rich and non-polyphenol-rich cranberry fruit extracts and their chemopreventive effects on colitis-associated colon cancer, J. Agric. Food Chem. 68 (2020) 6845-6853.
    [37]
    H.M.I. Abdallah, N.M. Ammar, M.F. Abdelhameed, et al., Protective mechanism of Acacia saligna butanol extract and its nano-formulations against ulcerative colitis in rats as revealed via biochemical and metabolomic assays, Biology 9 (2020), 195.
    [38]
    M. Lubis, G.A. Siregar, H. Bangun, et al., The effect of Roselle flower petals extract (Hibiscus sabdariffa Linn.) on reducing inflammation in dextran sodium sulfate induced colitis, Med. Glas. 17 (2020) 395-401.
    [39]
    S.H. Cestari, J.K. Bastos, L.C. Di Stasi, Intestinal anti-inflammatory activity of Baccharis dracunculifolia in the trinitrobenzenesulphonic acid model of rat colitis, Evid. Based Complement. Altern. Med. 2011 (2011) 1-9.
    [40]
    Y. Liu, L. Luo, Y. Luo, et al., Prebiotic properties of green and dark tea contribute to protective effects in chemical-induced colitis in mice: a fecal microbiota transplantation study, J. Agric. Food Chem. 68 (2020) 6368-6380.
    [41]
    T. Ortiz, F. Arguelles-Arias, M. Illanes, et al., Polyphenolic maqui extract as a potential nutraceutical to treat TNBS-induced Crohn's disease by the regulation of antioxidant and anti-inflammatory pathways, Nutrients 12 (2020), 1752.
    [42]
    J. Song, S.H. Shin, M.H. Park, et al., Fresh Saengshik showed a positive effect on mitigating dextran sulfate sodium-induced experimental colitis in mice, J. Med. Food 23 (2020) 459-464.
    [43]
    L.H. Maurer, C.B.B. Cazarin, A. Quatrin, et al., Grape peel powder attenuates the inflammatory and oxidative response of experimental colitis in rats by modulating the NF-κB pathway and activity of antioxidant enzymes, Nutr. Res. 76 (2020) 52-70.
    [44]
    G.C. Pistol, D.E. Marin, M.C. Rotar, et al., Bioactive compounds from dietary whole grape seed meal improved colonic inflammation via inhibition of MAPKs and NF-κB signaling in pigs with DSS induced colitis, J. Funct.Foods 66 (2020), 103708.
    [45]
    H. Ismaeil, W. Abdo, S. Amer, et al., Ameliorative effect of heat-killed Lactobacillus plantarum L.137 and/or Aloe vera against colitis in mice, Processes 8 (2020), 225.
    [46]
    M.E. El-Naggar, J. Hussein, S.M. El-sayed, et al., Protective effect of the functional yogurt based on Malva parviflora leaves extract nanoemulsion on acetic acid-induced ulcerative colitis in rats, J. Mater. Res. Technol. 9 (2020) 14500-14508.
    [47]
    J. Rocha, R. Direito, A. Lima, et al., Reduction of inflammation and colon injury by a Pennyroyal phenolic extract in experimental inflammatory bowel disease in mice, Biomed. Pharmacother. 118 (2019), 109351.
    [48]
    M.S. Stan, S.N. Voicu, S. Caruntu, et al., Antioxidant and anti-inflammatory properties of a Thuja occidentalis mother tincture for the treatment of ulcerative colitis, Antioxidants 8 (2019), 416.
    [49]
    D. Graf, J.M. Monk, D. Lepp, et al., Cooked red lentils dose-dependently modulate the colonic microenvironment in healthy C57Bl/6 male mice, Nutrients 11 (2019), 1853.
    [50]
    S. Babitha, K. Bindu, T. Nageena, et al., Fresh fruit juice of Opuntia dillenii haw. attenuates acetic acid-induced ulcerative colitis in rats, J. Diet. Suppl. 16 (2019) 431-442.
    [51]
    T. Shinde, A.P. Perera, R. Vemuri, et al., Synbiotic supplementation containing whole plant sugar cane fibre and probiotic spores potentiates protective synergistic effects in mouse model of IBD, Nutrients 11 (2019), 818.
    [52]
    W. Lee, Y.T. Tung, C.C. Wu, et al., Camellia oil (Camellia oleifera Abel.) modifies the composition of gut microbiota and alleviates acetic acid-induced colitis in rats, J. Agric. Food Chem. 66 (2018) 7384-7392.
    [53]
    Q.K. Alabi, R.O. Akomolafe, J.G. Omole, et al., Polyphenol-rich extract of Ocimum gratissimum leaves ameliorates colitis via attenuating colonic mucosa injury and regulating pro-inflammatory cytokines production and oxidative stress, Biomed. Pharmacother. 103 (2018) 812-822.
    [54]
    O.O. Abiodun, A.S. Sosanya, N. Nwadike, et al., Beneficial effect of Bidens pilosa L. (Asteraceae) in a rat model of colitis, J. Basic Clin. Physiol. Pharmacol. 31 (2020).
    [55]
    S. Periasamy, W.H. Wu, S. Chien, et al., Dietary Ziziphus jujuba fruit attenuates colitis-associated tumorigenesis: a pivotal role of the NF-κB/IL-6/JAK1/STAT3 pathway, Nutr. Cancer 72 (2020) 120-132.
    [56]
    M. Zhang, E. Viennois, M. Prasad, et al., Edible ginger-derived nanoparticles: a novel therapeutic approach for the prevention and treatment of inflammatory bowel disease and colitis-associated cancer, Biomaterials 101 (2016) 321-340.
    [57]
    N.H. Kim, S.M. Lee, Y.N. Kim, et al., Standardized fraction of Turbinaria ornata alleviates dextran sulfate sodium-induced chronic colitis in C57BL/6 mice via upregulation of FOXP3+ regulatory T cells, Biomolecules 10 (2020), 1463.
    [58]
    M.I. Yatoo, A. Gopalakrishnan, A. Saxena, et al., Anti-inflammatory drugs and herbs with special emphasis on herbal medicines for countering inflammatory diseases and disorders - a review, Recent Pat. Inflamm. Allergy Drug Discov. 12 (2018) 39-58.
    [59]
    J. Rocha, R. Leandro, R. Direito, et al., Attenuation of colonic injury and inflammation by administration of a phenolic extract of summer savory (Satureja hortensis L.) in experimental inflammatory bowel disease in mice, Appl. Sci. 10 (2020), 8465.
    [60]
    P.O.A. de Assis, G.C.B. Guerra, D.F. de Souza Araujo, et al., Intestinal anti-inflammatory activity of xique-xique (Pilosocereus gounellei A. Weber ex K. Schum. Bly. Ex Rowl) juice on acetic acid-induced colitis in rats, Food Funct. 10 (2019) 7275-7290.
    [61]
    X. Chen, M. Li, D. Li, et al., Ethanol extract of Pycnoporus sanguineus relieves the dextran sulfate sodium-induced experimental colitis by suppressing helper T cell-mediated inflammation via apoptosis induction, Biomed. Pharmacother. 127 (2020), 110212.
    [62]
    Y. Lin, W. Xiong, S. Xiao, et al., Pharmacoproteomics reveals the mechanism of Chinese dragon's blood in regulating the RSK/TSC2/mTOR/ribosome pathway in alleviation of DSS-induced acute ulcerative colitis, J. Ethnopharmacol. 263 (2020), 113221.
    [63]
    K. Kusmardi, D. Shavera, A. Estuningtyas, et al., The effect of mahkota dewa (Phaleria macrocarpa) (scheff.) fruit pericarp extract on inos in mice colon intermittently-induced by dextran sodium sulfate, Asian J. Pharm. Clin. Res. 10 (2017), 309.
    [64]
    C.B.B. Cazarin, A. Rodriguez-Nogales, F. Algieri, et al., Intestinal anti-inflammatory effects of Passiflora edulis peel in the dextran sodium sulphate model of mouse colitis, J. Funct.Foods 26 (2016) 565-576.
    [65]
    G. Orso, M.M. Solovyev, S. Facchiano, et al., Chestnut shell tannins: effects on intestinal inflammation and dysbiosis in zebrafish, Animals 11 (2021), 1538.
    [66]
    Y.N. Dey, G. Sharma, M.M. Wanjari, et al., Beneficial effect of Amorphophallus paeoniifolius tuber on experimental ulcerative colitis in rats, Pharm. Biol. 55 (2017) 53-62.
    [67]
    S. Jedidi, H. Sammari, H. Selmi, et al., Strong protective effects of Salvia officinalis L. leaves decoction extract against acetic acid-induced ulcerative colitis and metabolic disorders in rat, J. Funct.Foods 79 (2021), 104406.
    [68]
    G.R. de Morais Lima, F.D.F. Machado, L.L. Perico, et al., Anti-inflammatory intestinal activity of Combretum duarteanum Cambess. in trinitrobenzene sulfonic acid colitis model, World J. Gastroenterol. 23 (2017), 1353.
    [69]
    M. Saiyed, P. Sachdeva, M. Kukkar, Effectiveness of Ricinus communis root extract against dextran sodium sulfate induced ulcerative colitis in rats, Int. J. Green Pharm. 11 (2017) 84-91.
    [70]
    J.P. Veenstra, B. Vemu, R. Tocmo, et al., Pharmacokinetic analysis of carnosic acid and carnosol in standardized rosemary extract and the effect on the disease activity index of DSS-induced colitis, Nutrients 13 (2021), 773.
    [71]
    S. Li, T. Wang, B. Wu, et al., Anthocyanin-containing purple potatoes ameliorate DSS-induced colitis in mice, J. Nutr. Biochem. 93 (2021), 108616.
    [72]
    Y. Wang, H. Tao, H. Huang, et al., The dietary supplement Rhodiola crenulata extract alleviates dextran sulfate sodium-induced colitis in mice through anti-inflammation, mediating gut barrier integrity and reshaping the gut microbiome, Food Funct. 12 (2021) 3142-3158.
    [73]
    J. Li, M. Li, K. Ye, et al., Chemical profile of Xian-He-Cao-Chang-Yan formula and its effects on ulcerative colitis, J. Ethnopharmacol. 267 (2021), 113517.
    [74]
    S.H. Kang, Y.J. Song, Y.D. Jeon, et al., Comparative study of anti-inflammatory effect on DSS-induced ulcerative colitis between novel Glycyrrhiza variety and official compendia, Appl. Sci. 11 (2021), 1545.
    [75]
    N. Tatiya-aphiradee, W. Chatuphonprasert, K. Jarukamjorn, Ethanolic Garcinia mangostana extract and α-mangostin improve dextran sulfate sodium-induced ulcerative colitis via the suppression of inflammatory and oxidative responses in ICR mice, J. Ethnopharmacol. 265 (2021), 113384.
    [76]
    N. Pengkumsri, B.S. Sivamaruthi, S. Sirilun, et al., Dietary supplementation of Thai black rice bran extract and yeast beta-glucan protects the dextran sodium sulphate mediated colitis induced rat, RSC Adv. 7 (2017) 396-402.
    [77]
    H. Kim, N. Banerjee, R.C. Barnes, et al., Mango polyphenolics reduce inflammation in intestinal colitis: involvement of the miR-126/PI3K/AKT/mTOR axis in vitro and in vivo, Mol. Carcinog. 56 (2017) 197-207.
    [78]
    Z. Zhang, J. Liu, P. Shen, et al., Zanthoxylum bungeanum pericarp extract prevents dextran sulfate sodium-induced experimental colitis in mice via the regulation of TLR4 and TLR4-related signaling pathways, Int. Immunopharmacol. 41 (2016) 127-135.
    [79]
    J.H. Kim, Y.S. Won, H.D. Cho, et al., Protective effect of Prunus mume fermented with mixed lactic acid bacteria in dextran sodium sulfate-induced colitis, Foods 10 (2020), 58.
    [80]
    R.A. El-Shiekh, D. Hussein, A.H. Atta, et al., Anti-inflammatory activity of Jasminum grandiflorum L. subsp. floribundum (Oleaceae) in inflammatory bowel disease and arthritis models, Biomed. Pharmacother. 140 (2021), 111770.
    [81]
    K.H. Han, J.M. Park, M. Jeong, et al., Heme oxygenase-1 induction and anti-inflammatory actions of Atractylodes macrocephala and Taraxacum herba extracts prevented colitis and was more effective than sulfasalazine in preventing relapse, Gut Liver 11 (2017) 655-666.
    [82]
    J.H. Choi, K.S. Chung, B.R. Jin, et al., Anti-inflammatory effects of an ethanol extract of Aster glehni via inhibition of NF-κB activation in mice with DSS-induced colitis, Food Funct. 8 (2017) 2611-2620.
    [83]
    J.Y. Cho, H.Y. Kim, H.M. Kim, et al., Standardized ethanolic extract of the rhizome of Curcuma xanthorrhiza prevents murine ulcerative colitis by regulation of inflammation, J. Funct.Foods 30 (2017) 282-289.
    [84]
    R. Li, M.H. Kim, A.K. Sandhu, et al., Muscadine grape (Vitis rotundifolia) or wine phytochemicals reduce intestinal inflammation in mice with dextran sulfate sodium-induced colitis, J. Agric. Food Chem. 65 (2017) 769-776.
    [85]
    B. Wang, Y. Li, M. Mizu, et al., Protective effect of sugar cane extract against dextran sulfate sodium-induced colonic inflammation in mice, Tissue Cell 49 (2017) 8-14.
    [86]
    J.Y. Hong, K.S. Chung, J.S. Shin, et al., Anti-colitic effects of ethanol extract of Persea americana mill. through suppression of pro-inflammatory mediators via NF-κB/STAT3 inactivation in dextran sulfate sodium-induced colitis mice, Int. J. Mol. Sci. 20 (2019), 177.
    [87]
    M.R. Akanda, H.H. Nam, W. Tian, et al., Regulation of JAK2/STAT3 and NF-κB signal transduction pathways; Veronica polita alleviates dextran sulfate sodium-induced murine colitis, Biomed. Pharmacother. 100 (2018) 296-303.
    [88]
    M.G. Helal, A.G. Abd Elhameed, Graviola mitigates acetic acid-induced ulcerative colitis in rats: Insight on apoptosis and Wnt/Hh signaling crosstalk, Environ. Sci. Pollut. Res. 28 (2021) 29615-29628.
    [89]
    A.W.L. Andrade, G.C.B. Guerra, D.F. de Souza Araujo, et al., Anti-inflammatory and chemopreventive effects of Bryophyllum pinnatum (lamarck) leaf extract in experimental colitis models in rodents, Front. Pharmacol. 11 (2020), 998.
    [90]
    C.F. Adjouzem, A. Gilbert, M. Mbiantcha, et al., Effects of aqueous and methanolic extracts of stem bark of Alstonia boonei de wild. (Apocynaceae) on dextran sodium sulfate-induced ulcerative colitis in wistar rats, Evid. Based Complement. Alternat. Med. 2020 (2020), 4918453.
    [91]
    V. da Silva, A. de Araujo, D. Araujo, et al., Intestinal anti-inflammatory activity of the aqueous extract from Ipomoea asarifolia in DNBS-induced colitis in rats, Int. J. Mol. Sci. 19 (2018), 4016.
    [92]
    B. Qian, C. Wang, Z. Zeng, et al., Ameliorative effect of sinapic acid on dextran sodium sulfate- (DSS-) induced ulcerative colitis in Kunming (KM) mice, Oxid. Med. Cell. Longev. 2020 (2020) 1-13.
    [93]
    M.M. Tambuwala, M.N. Khan, P. Thompson, et al., Albumin nano-encapsulation of caffeic acid phenethyl ester and piceatannol potentiated its ability to modulate HIF and NF-κB pathways and improves therapeutic outcome in experimental colitis, Drug Deliv. Transl. Res. 9 (2019) 14-24.
    [94]
    F. Zhang, N. Ma, Y. Gao, et al., Therapeutic effects of 6-gingerol, 8-gingerol, and 10-gingerol on dextran sulfate sodium-induced acute ulcerative colitis in rats, Phytother. Res. 31 (2017) 1427-1432.
    [95]
    Y. Dong, Q. Hou, J. Lei, et al., Quercetin alleviates intestinal oxidative damage induced by H2O2 via modulation of GSH: in vitro screening and in vivo evaluation in a colitis model of mice, ACS Omega 5 (2020) 8334-8346.
    [96]
    E. Pagano, B. Romano, F.A. Iannotti, et al., The non-euphoric phytocannabinoid cannabidivarin counteracts intestinal inflammation in mice and cytokine expression in biopsies from UC pediatric patients, Pharmacol. Res. 149 (2019), 104464.
    [97]
    J.B. Park, D.W. Kim, K.T. Lim, et al., A 75 kDa glycoprotein isolated from Cudrania tricuspidata Bureau induces colonic epithelial proliferation and ameliorates mouse colitis induced by dextran sulfate sodium, Chin. J. Nat. Med. 19 (2021) 46-55.
    [98]
    L. Luo, J. Yan, B. Chen, et al., The effect of menthol supplement diet on colitis-induced colon tumorigenesis and intestinal microbiota, Am. J. Transl. Res. 13 (2021) 38-56.
    [99]
    S. Momtaz, M. Navabakhsh, N. Bakouee, et al., Cinnamaldehyde targets TLR-4 and inflammatory mediators in acetic-acid induced ulcerative colitis model, Biologia 76 (2021) 1817-1827.
    [100]
    N.F. Sadek, N.D. Yuliana, E. Prangdimurti, et al., Plant sterol esters in extruded food model inhibits colon carcinogenesis by suppressing inflammation and stimulating apoptosis, J. Med. Food 20 (2017) 659-666.
    [101]
    M. Gu, S. Pan, Q. Li, et al., Chitosan and chitooligosaccharides attenuate soyabean meal-induced intestinal inflammation of turbot (Scophthalmus maximus): possible involvement of NF-κB, activator protein-1 and mitogen-activated protein kinases pathways, Br. J. Nutr. 126 (2021) 1651-1662.
    [102]
    V. Raj, B. Venkataraman, S. Almarzooqi, et al., Nerolidol mitigates colonic inflammation: an experimental study using both in vivo and in vitro models, Nutrients 12 (2020), 2032.
    [103]
    M.U. Rehman, I.A. Rather, Myricetin abrogates cisplatin-induced oxidative stress, inflammatory response, and goblet cell disintegration in colon of wistar rats, Plants 9 (2019), 28.
    [104]
    G. Guo, W. Shi, F. Shi, et al., Anti-inflammatory effects of eriocitrin against the dextran sulfate sodium-induced experimental colitis in murine model, J. Biochem. Mol. Toxicol. 33 (2019), e22400.
    [105]
    I. Sukhotnik, D. Moati, R. Shaoul, et al., Quercetin prevents small intestinal damage and enhances intestinal recovery during methotrexate-induced intestinal mucositis of rats, Food Nutr. Res. 62 (2018).
    [106]
    Y. Yu, C. Zheng, X. Lu, et al., GB1a ameliorates ulcerative colitis via regulation of the NF-κB and Nrf2 signaling pathways in an experimental model, Front. Med. 8 (2021) 654867.
    [107]
    C.W. Cho, S. Ahn, T.G. Lim, et al., Cynanchum wilfordii polysaccharides suppress dextran sulfate sodium-induced acute colitis in mice and the production of inflammatory mediators from macrophages, Mediat. Inflamm. 2017 (2017) 1-14.
    [108]
    D. Liu, X. Yu, H. Sun, et al., Flos lonicerae flavonoids attenuate experimental ulcerative colitis in rats via suppression of NF-κB signaling pathway, Naunyn Schmiedebergs Arch. Pharmacol. 393 (2020) 2481-2494.
    [109]
    X. Zhao, H. Liu, Y. Wu, et al., Intervention with the crude polysaccharides of Physalis pubescens L. mitigates colitis by preventing oxidative damage, aberrant immune responses, and dysbacteriosis, J. Food Sci. 85 (2020) 2596-2607.
    [110]
    X. Huo, D. Liu, L. Gao, et al., Flavonoids extracted from licorice prevents colitis-associated carcinogenesis in AOM/DSS mouse model, Int. J. Mol. Sci. 17 (2016), 1343.
    [111]
    X. Xia, L. Zhu, Z. Lei, et al., Feruloylated oligosaccharides alleviate dextran sulfate sodium-induced colitis in vivo, J. Agric. Food Chem. 67 (2019) 9522-9531.
    [112]
    S. Riemschneider, M. Hoffmann, U. Slanina, et al., Indol-3-carbinol and quercetin ameliorate chronic DSS-induced colitis in C57BL/6 mice by AhR-mediated anti-inflammatory mechanisms, Int. J. Environ. Res. Public Health. 18 (2021), 2262.
    [113]
    X. Wu, M. Song, Z. Gao, et al., Nobiletin and its colonic metabolites suppress colitis-associated colon carcinogenesis by down-regulating iNOS, inducing antioxidative enzymes and arresting cell cycle progression, J. Nutr. Biochem. 42 (2017) 17-25.
    [114]
    F. Biasi, G. Leonarduzzi, P.I. Oteiza, et al., Inflammatory bowel disease: mechanisms, redox considerations, and therapeutic targets, Antioxid. Redox Signal. 19 (2013) 1711-1747.
    [115]
    M. Kasembeli, U. Bharadwaj, P. Robinson, et al., Contribution of STAT3 to inflammatory and fibrotic diseases and prospects for its targeting for treatment, Int. J. Mol. Sci. 19 (2018), 2299.
    [116]
    Q. Wang, N. Huber, G. Noel, et al., NF-κΒ inhibition is ineffective in blocking cytokine-induced IL-8 production but P38 and STAT1 inhibitors are effective, Inflamm. Res. 61 (2012) 977-985.
    [117]
    I. Atreya, M.F. Neurath, Immune cells in colorectal cancer: prognostic relevance and therapeutic strategies, Expert Rev. Anticancer Ther. 8 (2008) 561-572.
    [118]
    F.J. Quintana, A.S. Basso, A.H. Iglesias, et al., Control of Treg and TH17 cell differentiation by the aryl hydrocarbon receptor, Nature 453 (2008) 65-71.
    [119]
    M.J. Tosiek, L. Fiette, S. El Daker, et al., IL-15-dependent balance between Foxp3 and RORγt expression impacts inflammatory bowel disease, Nat. Commun. 7 (2016), 10888.
    [120]
    M.M. Gaschler, B.R. Stockwell, Lipid peroxidation in cell death, Biochem. Biophys. Res. Commun. 482 (2017) 419-425.
    [121]
    K. Danis-Wlodarczyk, D. Vandenheuvel, H.B. Jang, et al., A proposed integrated approach for the preclinical evaluation of phage therapy in Pseudomonas infections, Sci. Rep. 6 (2016), 28115.
    [122]
    J.H. Sellin, Y. Wang, P. Singh, et al., β-Catenin stabilization imparts crypt progenitor phenotype to hyperproliferating colonic epithelia, Exp. Cell Res. 315 (2009) 97-109.
    [123]
    S. Umar, Y. Wang, A.P. Morris, et al., Dual alterations in casein kinase I-ε and GSK-3β modulate β-catenin stability in hyperproliferating colonic epithelia, Am. J. Physiol. Gastrointest. Liver Physiol. 292 (2007) G599-G607.
    [124]
    I. Soufli, R. Toumi, H. Rafa, et al., Overview of cytokines and nitric oxide involvement in immuno-pathogenesis of inflammatory bowel diseases, World J. Gastrointest. Pharmacol. Ther. 7 (2016) 353-360.
    [125]
    Y. Chang, L. Zhai, J. Peng, et al., Phytochemicals as regulators of Th17/Treg balance in inflammatory bowel diseases, Biomed. Pharmacother. 141 (2021), 111931.
    [126]
    M. Nikkhah-Bodaghi, I. Maleki, S. Agah, et al., Zingiber officinale and oxidative stress in patients with ulcerative colitis: a randomized, placebo-controlled, clinical trial, Complement. Ther. Med. 43 (2019) 1-6.
    [127]
    A.G. Long, E.T. Lundsmith, K.E. Hamilton, Inflammation and colorectal cancer, Curr. Colorectal Cancer Rep. 13 (2017) 341-351.
    [128]
    Y. Huang, J. Zhou, C. Qu, et al., Anti-inflammatory effects of Brucea javanica oil emulsion by suppressing NF-κB activation on dextran sulfate sodium-induced ulcerative colitis in mice, J. Ethnopharmacol. 198 (2017) 389-398.
    [129]
    H. Kawakami, M. Tomita, T. Matsuda, et al., Transcriptional activation of survivin through the NF-κB pathway by human T-cell leukemia virus type I tax, Int. J. Cancer 115 (2005) 967-974.
  • 加载中

Catalog

    通讯作者: 陈斌, bchen63@163.com
    • 1. 

      沈阳化工大学材料科学与工程学院 沈阳 110142

    1. 本站搜索
    2. 百度学术搜索
    3. 万方数据库搜索
    4. CNKI搜索

    Figures(1)

    Article Metrics

    Article views (207) PDF downloads(17) Cited by()
    Proportional views
    Related

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return