1
|
Darkazally AZ, Alnour A, Homsi S. Troxerutin effect on gastric ulcers induced by ketorolac in rats: Relation with oxidative stress. Heliyon 2024; 10:e38893. [PMID: 39435065 PMCID: PMC11492593 DOI: 10.1016/j.heliyon.2024.e38893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 09/03/2024] [Accepted: 10/01/2024] [Indexed: 10/23/2024] Open
Abstract
Gastric ulcers are an essential side effect associated with the use of non-steroidal anti-inflammatory drugs (NSAIDs). Free radicals are one of the important mechanisms contributing to the development of gastric ulcers caused by NSAIDs. This prompted us to choose troxerutin, which has antioxidant and anti-inflammatory effects, especially with a lack of studies investigating the preventive effect of troxerutin on gastric ulcers. Twenty-nine rats were divided into five groups: A Vehicle group, a Keto group (30 mg/kg of ketorolac), and two troxerutin groups (150 mg/kg or 200 mg/kg of troxerutin, respectively). A Miso group was used as a reference with (100 μg/kg of misoprostol). Troxerutin and misoprostol were administered orally 1 h before ketorolac. The ulcer index was determined considering the numbers and severity of ulcerations. Gastric tissue inflammation was evaluation microscopically. Both thiobarbituric acid reactive substance levels and catalase activity were measured as markers of oxidative stress in gastric tissue. Our data showed an improvement in ulcer indices with troxerutin and misoprostol compared with ketorolac, with improvement in gastric inflammation observed with misoprostol but not with troxerutin. These results were accompanied by a reduction in gastric oxidative stress induced by ketorolac with both troxerutin and misoprostol. This study highlights, for the first time, the antioxidant effect of troxerutin on gastric ulcers. This effect may contribute to the good prevention of ketorolac-induced gastric ulcers.
Collapse
Affiliation(s)
| | - Amirah Alnour
- Department of Histology and Pathological Anatomy, Faculty of Dentistry, Damascus University, Damascus, Syria
| | - Shadi Homsi
- Department of Pharmacology, Faculty of Pharmacy, Damascus University, Damascus, Syria
| |
Collapse
|
2
|
Singh S, Sharma S, Sharma H. Potential Impact of Bioactive Compounds as NLRP3 Inflammasome Inhibitors: An Update. Curr Pharm Biotechnol 2024; 25:1719-1746. [PMID: 38173061 DOI: 10.2174/0113892010276859231125165251] [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: 09/02/2023] [Revised: 10/22/2023] [Accepted: 10/26/2023] [Indexed: 01/05/2024]
Abstract
The inflammasome NLRP3 comprises a caspase recruitment domain, a pyrin domain containing receptor 3, an apoptosis-linked protein like a speck containing a procaspase-1, and an attached nucleotide domain leucine abundant repeat. There are a wide variety of stimuli that can activate the inflammasome NLRP3. When activated, the protein NLRP3 appoints the adapter protein ASC. Adapter ASC protein then recruits the procaspase-1 protein, which causes the procaspase- 1 protein to be cleaved and activated, which induces cytokines. At the same time, abnormal activation of inflammasome NLRP3 is associated with many diseases, such as diabetes, atherosclerosis, metabolic syndrome, cardiovascular and neurodegenerative diseases. As a result, a significant amount of effort has been put into comprehending the mechanisms behind its activation and looking for their specific inhibitors. In this review, we primarily focused on phytochemicals that inhibit the inflammasome NLRP3, as well as discuss the defects caused by NLRP3 signaling. We conducted an in-depth research review by searching for relevant articles in the Scopus, Google Scholar, and PubMed databases. By gathering information on phytochemical inhibitors that block NLRP3 inflammasome activation, a complicated balance between inflammasome activation or inhibition with NLRP3 as a key role was revealed in NLRP3-driven clinical situations.
Collapse
Affiliation(s)
- Sonia Singh
- Department of Pharmacy, Institute of Pharmaceutical Research, GLA University, Uttar Pradesh-281406, India
| | - Shiwangi Sharma
- Department of Pharmacy, Institute of Pharmaceutical Research, GLA University, Uttar Pradesh-281406, India
| | - Himanshu Sharma
- Department of Computer Engineering & Applications, GLA University, Uttar Pradesh-281406, India
| |
Collapse
|
3
|
He Y, DeBenedictis JN, Caiment F, van Breda SGJ, de Kok TMCM. Analysis of cell-specific transcriptional responses in human colon tissue using CIBERSORTx. Sci Rep 2023; 13:18281. [PMID: 37880448 PMCID: PMC10600214 DOI: 10.1038/s41598-023-45582-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 10/21/2023] [Indexed: 10/27/2023] Open
Abstract
Diet is an important determinant of overall health, and has been linked to the risk of various cancers. To understand the mechanisms involved, transcriptomic responses from human intervention studies are very informative. However, gene expression analysis of human biopsy material only represents the average profile of a mixture of cell types that can mask more subtle, but relevant cell-specific changes. Here, we use the CIBERSORTx algorithm to generate single-cell gene expression from human multicellular colon tissue. We applied the CIBERSORTx to microarray data from the PHYTOME study, which investigated the effects of different types of meat on transcriptional and biomarker changes relevant to colorectal cancer (CRC) risk. First, we used single-cell mRNA sequencing data from healthy colon tissue to generate a novel signature matrix in CIBERSORTx, then we determined the proportions and gene expression of each separate cell type. After comparison, cell proportion analysis showed a continuous upward trend in the abundance of goblet cells and stem cells, and a continuous downward trend in transit amplifying cells after the addition of phytochemicals in red meat products. The dietary intervention influenced the expression of genes involved in the growth and division of stem cells, the metabolism and detoxification of enterocytes, the translation and glycosylation of goblet cells, and the inflammatory response of innate lymphoid cells. These results show that our approach offers novel insights into the heterogeneous gene expression responses of different cell types in colon tissue during a dietary intervention.
Collapse
Affiliation(s)
- Yueqin He
- Department of Toxicogenomics, GROW - School for Oncology and Reproduction, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, P.O. Box 616, 6200 MD, Maastricht, The Netherlands.
| | - Julia Nicole DeBenedictis
- Department of Toxicogenomics, GROW - School for Oncology and Reproduction, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Florian Caiment
- Department of Toxicogenomics, GROW - School for Oncology and Reproduction, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Simone G J van Breda
- Department of Toxicogenomics, GROW - School for Oncology and Reproduction, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Theo M C M de Kok
- Department of Toxicogenomics, GROW - School for Oncology and Reproduction, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| |
Collapse
|
4
|
He Y, Zheng J, Ye B, Dai Y, Nie K. Chemotherapy-induced gastrointestinal toxicity: Pathogenesis and current management. Biochem Pharmacol 2023; 216:115787. [PMID: 37666434 DOI: 10.1016/j.bcp.2023.115787] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 09/06/2023]
Abstract
Chemotherapy is the most common treatment for malignant tumors. However, chemotherapy-induced gastrointestinal toxicity (CIGT) has been a major concern for cancer patients, which reduces their quality of life and leads to treatment intolerance and even cessation. Nevertheless, prevention and treatment for CIGT are challenging, due to the prevalence and complexity of the condition. Chemotherapeutic drugs directly damage gastrointestinal mucosa to induce CIGT, including nausea, vomiting, anorexia, gastrointestinal mucositis, and diarrhea, etc. The pathogenesis of CIGT involves multiple factors, such as gut microbiota disorders, inflammatory responses and abnormal neurotransmitter levels, that synergistically contribute to its occurrence and development. In particular, the dysbiosis of gut microbiota is usually linked to abnormal immune responses that increases inflammatory cytokines' expression, which is a common characteristic of many types of CIGT. Chemotherapy-induced intestinal neurotoxicity is also a vital concern in CIGT. Currently, modern medicine is the dominant treatment of CIGT, however, traditional Chinese medicine (TCM) has attracted interest as a complementary and alternative therapy that can greatly alleviate CIGT. Accordingly, this review aimed to comprehensively summarize the pathogenesis and current management of CIGT using PubMed and Google Scholar databases, and proposed that future research for CIGT should focus on the gut microbiota, intestinal neurotoxicity, and promising TCM therapies, which may help to develop more effective interventions and optimize managements of CIGT.
Collapse
Affiliation(s)
- Yunjing He
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jingrui Zheng
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Binbin Ye
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yongzhao Dai
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Ke Nie
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
5
|
da Silva KS, Abboud KY, Schiebel CS, de Oliveira NMT, Bueno LR, de Mello Braga LLV, da Silveira BC, Santos IWFD, Gomes EDS, Gois MB, Cordeiro LMC, Maria Ferreira D. Polysaccharides from Passion Fruit Peels: From an Agroindustrial By-Product to a Viable Option for 5-FU-Induced Intestinal Damage. Pharmaceuticals (Basel) 2023; 16:912. [PMID: 37513823 PMCID: PMC10383750 DOI: 10.3390/ph16070912] [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: 05/17/2023] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 07/30/2023] Open
Abstract
Gastrointestinal mucositis is a serious and dose-limiting toxic side effect of oncologic treatment. Interruption of cancer treatment due to gastrointestinal mucositis leads to a significant decrease in cure rates and consequently to the deterioration of a patient's quality of life. Natural polysaccharides show a variety of beneficial effects, including a gastroprotective effect. Treatment with soluble dietary fiber (SDF) from yellow passion fruit (Passiflora edulis) biomass residues protected the gastric and intestinal mucosa in models of gastrointestinal injury. In this study, we investigated the protective therapeutic effect of SDF on 5-FU-induced mucositis in male and female mice. Oral treatment of the animals with SDF did not prevent weight loss but reduced the disease activity index and preserved normal intestinal function by alleviating diarrhea and altered gastrointestinal transit. SDF preserved the length of the colon and histological damage caused by 5-FU. SDF significantly restored the oxidative stress and inflammation in the intestine and the enlargement and swelling of the spleen induced by 5-FU. In conclusion, SDF may be a promising adjuvant strategy for the prevention and treatment of intestinal mucositis induced by 5-FU.
Collapse
Affiliation(s)
- Karien Sauruk da Silva
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba 80250-200, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim No 1532, Curitiba 80250-200, Brazil
| | - Kahlile Youssef Abboud
- Department of Biochemistry and Molecular Biology, Federal University of Paraná, Curitiba 81531-980, Brazil
| | - Carolina Silva Schiebel
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba 80250-200, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim No 1532, Curitiba 80250-200, Brazil
| | - Natalia Mulinari Turin de Oliveira
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba 80250-200, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim No 1532, Curitiba 80250-200, Brazil
| | - Laryssa Regis Bueno
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba 80250-200, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim No 1532, Curitiba 80250-200, Brazil
| | - Lara Luisa Valerio de Mello Braga
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba 80250-200, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim No 1532, Curitiba 80250-200, Brazil
| | - Bruna Carla da Silveira
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba 80250-200, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim No 1532, Curitiba 80250-200, Brazil
| | - Isabella Wzorek França Dos Santos
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba 80250-200, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim No 1532, Curitiba 80250-200, Brazil
| | - Everton Dos Santos Gomes
- Programa de Pós-Graduação em Imunologia, Universidade Federal da Bahia, Salvador 40231-300, Brazil
- Programa de Pós-Graduação em Biociências e Saúde, Universidade Federal de Rondonópolis, Rondonópolis 78736-900, Brazil
| | - Marcelo Biondaro Gois
- Programa de Pós-Graduação em Imunologia, Universidade Federal da Bahia, Salvador 40231-300, Brazil
- Programa de Pós-Graduação em Biociências e Saúde, Universidade Federal de Rondonópolis, Rondonópolis 78736-900, Brazil
| | | | - Daniele Maria Ferreira
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba 80250-200, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim No 1532, Curitiba 80250-200, Brazil
| |
Collapse
|
6
|
Dahlgren D, Lennernäs H. Review on the effect of chemotherapy on the intestinal barrier: Epithelial permeability, mucus and bacterial translocation. Biomed Pharmacother 2023; 162:114644. [PMID: 37018992 DOI: 10.1016/j.biopha.2023.114644] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/30/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
Chemotherapy kills fast-growing cells including gut stem cells. This affects all components of the physical and functional intestinal barrier, i.e., the mucus layer, epithelium, and immune system. This results in an altered intestinal permeability of toxic compounds (e.g., endotoxins) as well as luminal bacterial translocation into the mucosa and central circulation. However, there is uncertainty regarding the relative contributions of the different barrier components for the development of chemotherapy-induced gut toxicity. This review present an overview of the intestinal mucosal barrier determined with various types of molecular probes and methods, and how they are affected by chemotherapy based on reported rodent and human data. We conclude that there is overwhelming evidence that chemotherapy increases bacterial translocation, and that it affects the mucosal barrier by rendering the mucosa more permeable to large permeability probes. Chemotherapy also seems to impede the intestinal mucus barrier, even though this has been less clearly evaluated from a functional standpoint but certainly plays a role in bacteria translocation. Combined, it is however difficult to outline a clear temporal or succession between the different gastrointestinal events and barrier functions, especially as chemotherapy-induced neutropenia is also involved in intestinal immunological homeostasis and bacterial translocation. A thorough characterization of this would need to include a time dependent development of neutropenia, intestinal permeability, and bacterial translocation, ideally after a range of chemotherapeutics and dosing regimens.
Collapse
|
7
|
Luisa Valerio de Mello Braga L, Simão G, Silva Schiebel C, Caroline Dos Santos Maia A, Mulinari Turin de Oliveira N, Barbosa da Luz B, Rita Corso C, Soares Fernandes E, Maria Ferreira D. Rodent models for anticancer toxicity studies: contributions to drug development and future perspectives. Drug Discov Today 2023:103626. [PMID: 37224998 DOI: 10.1016/j.drudis.2023.103626] [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: 08/12/2022] [Revised: 05/08/2023] [Accepted: 05/17/2023] [Indexed: 05/26/2023]
Abstract
Antineoplastic treatment induces a type of gastrointestinal toxicity known as mucositis. Findings in animal models are usually easily reproducible, and standardized treatment regimens are often used, thus supporting translational science. Essential characteristics of mucositis, including intestinal permeability, inflammation, immune and oxidative responses, and tissue repair mechanisms, can be easily investigated in these models. Given the effects of mucositis on the quality of life of patients with cancer, and the importance of experimental models in the development of more effective new therapeutic alternatives, this review discusses progress and current challenges in using experimental models of mucositis in translational pharmacology research. Teaser Experimental models for studying gastrointestinal mucositis have provided a wealth of information improving the understanding of antineoplastic toxicity.
Collapse
Affiliation(s)
- Lara Luisa Valerio de Mello Braga
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil; Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Gisele Simão
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil; Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Carolina Silva Schiebel
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil; Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Andressa Caroline Dos Santos Maia
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil; Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Natalia Mulinari Turin de Oliveira
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil; Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Bruna Barbosa da Luz
- Departamento de Farmacologia, Universidade Federal do Paraná, Curitiba, PR, Brazil
| | - Claudia Rita Corso
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil; Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Elizabeth Soares Fernandes
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil; Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Daniele Maria Ferreira
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil; Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil.
| |
Collapse
|
8
|
Chen KJ, Huang YL, Kuo LM, Chen YT, Hung CF, Hsieh PW. Protective role of casuarinin from Melastoma malabathricum against a mouse model of 5-fluorouracil-induced intestinal mucositis: Impact on inflammation and gut microbiota dysbiosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 101:154092. [PMID: 35430483 DOI: 10.1016/j.phymed.2022.154092] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND 5-FU-induced intestinal mucositis (FUIIM) is a common gastrointestinal side effect of chemotherapy, leading to gastric pain in clinical cancer patients. In a previous study, we demonstrated that neutrophil elastase (NE) inhibitors could alleviate FUIIM and manipulate the homeostasis of the gut microbiota. The root of Melastoma malabathricum, also called Ye-Mu-Dan, has been used as a traditional Chinese medicine for gastrointestinal disease. Water extract of the roots of M. malabathricum exhibits an inhibitory effect on NE, with an IC50 value of 9.13 μg/ml. PURPOSE In this study, we aimed to isolate an anti-NE compound from the root of M. malabathricum and to determine the protective effect of the bioactive component on a mouse model of FUIIM with respect to tissue damage, inflammation, intestinal barrier dysfunction, and gut microbiota dysbiosis. METHODS A water extract of the roots of M. malabathricum was prepared and its major bioactive compound, was identified using bioactivity-guided fractionation. The effects of samples on the inhibition of NE activity were evaluated using enzymatic assays. To evaluate the effects of the bioactive compound in an FUIIM animal model, male C57BL/6 mice treated with or without casuarinin (50 and 100 mg/kg/day, p.o.), and then received of 5-fluorouracil (50 mg/kg/day) intraperitoneally for 5 days to induce FUIIM. Histopathological staining was used to monitor the tissue damage, proliferation of intestinal crypts, and expression of tight junction proteins. The inflammation score was estimated by determining the levels of oxidative stress, neutrophil-related proteases, and proinflammatory cytokines in tissue and serum. The ecology of the gut microbiota was evaluated using 16S rRNA gene sequencing. RESULTS Casuarinin had the most potent and selective effect against NE, with an IC50 value of 2.79 ± 0.07 μM. Casuarinin (100 mg/kg/day, p.o.) significantly improved 5-FU-induced body weight loss together with food intake reduction, and it also significantly reversed villus atrophy, restored the proliferative activity of the intestinal crypts, and suppressed inflammation and intestinal barrier dysfunction in the mouse model of FUIIM. Casuarinin also reversed 5-FU-induced gut microbiota dysbiosis, particularly the abundance of Actinobacteria, Candidatus Arthromitus, and Lactobacillus murinus, and the Firmicutes-to-Bacteroidetes ratio. CONCLUSION This study firstly showed that casuarinin isolated from the root part of M. malabathricum could be used as a NE inhibitor, whereas it could improve FUIIM by modulating inflammation, intestinal barrier dysfunction, and gut microbiota dysbiosis. In summary, exploring anti-NE natural product may provide a way to find candidate for improvement of FUIIM.
Collapse
Affiliation(s)
- Kung-Ju Chen
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Natural Products, School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan 333, Taiwan
| | - Yu-Ling Huang
- Graduate Institute of Natural Products, School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan 333, Taiwan
| | - Liang-Mou Kuo
- Department of General Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Yi-Ting Chen
- Graduate Institute of Natural Products, School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan 333, Taiwan
| | - Chi-Feng Hung
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan; Program in Pharmaceutical Biotechnology, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Pei-Wen Hsieh
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Natural Products, School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan 333, Taiwan; Department of General Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan; Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan, Taiwan.
| |
Collapse
|
9
|
The Intestinal Redox System and Its Significance in Chemotherapy-Induced Intestinal Mucositis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7255497. [PMID: 35585883 PMCID: PMC9110227 DOI: 10.1155/2022/7255497] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/04/2022] [Accepted: 04/09/2022] [Indexed: 12/12/2022]
Abstract
Chemotherapy-induced intestinal mucositis (CIM) is a significant dose-limiting adverse reaction brought on by the cancer treatment. Multiple studies reported that reactive oxygen species (ROS) is rapidly produced during the initial stages of chemotherapy, when the drugs elicit direct damage to intestinal mucosal cells, which, in turn, results in necrosis, mitochondrial dysfunction, and ROS production. However, the mechanism behind the intestinal redox system-based induction of intestinal mucosal injury and necrosis of CIM is still undetermined. In this article, we summarized relevant information regarding the intestinal redox system, including the composition and regulation of redox enzymes, ROS generation, and its regulation in the intestine. We innovatively proposed the intestinal redox “Tai Chi” theory and revealed its significance in the pathogenesis of CIM. We also conducted an extensive review of the English language-based literatures involving oxidative stress (OS) and its involvement in the pathological mechanisms of CIM. From the date of inception till July 31, 2021, 51 related articles were selected. Based on our analysis of these articles, only five chemotherapeutic drugs, namely, MTX, 5-FU, cisplatin, CPT-11, and oxaliplatin were shown to trigger the ROS-based pathological mechanisms of CIM. We also discussed the redox system-mediated modulation of CIM pathogenesis via elaboration of the relationship between chemotherapeutic drugs and the redox system. It is our belief that this overview of the intestinal redox system and its role in CIM pathogenesis will greatly enhance research direction and improve CIM management in the future.
Collapse
|
10
|
Arafah A, Rehman MU, Ahmad A, AlKharfy KM, Alqahtani S, Jan BL, Almatroudi NM. Myricetin (3,3',4',5,5',7-Hexahydroxyflavone) Prevents 5-Fluorouracil-Induced Cardiotoxicity. ACS OMEGA 2022; 7:4514-4524. [PMID: 35155943 PMCID: PMC8829927 DOI: 10.1021/acsomega.1c06475] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 01/12/2022] [Indexed: 05/05/2023]
Abstract
5-Fluorouracil (5-FU) is a strong anti-cancer drug used to manage numerous cancers. Cardiotoxicity, renal toxicity, and liver toxicity are some of the adverse effects which confine its clinical use to some extent. 5-FU-induced organ injuries are associated with redox imbalance, inflammation, and damage to heart functioning, particularly in the present study. Myricetin is an abundant flavonoid, commonly extracted from berries and herbs having anti-oxidative and anti-cancer activities. We planned the current work to explore the beneficial effects of myricetin against 5-FU-induced cardiac injury in Wistar rats through a biochemical and histological approach. Prophylactic myricetin treatment at two doses (25 and 50 mg/kg) was given to rats orally for 21 days against cardiac injury induced by a single injection of 5-FU (150 mg/kg b.wt.) given on the 20th day intraperitoneally. The 5-FU injection induced oxidative stress, inflammation, and extensive cardiac damage. Nevertheless, myricetin alleviated markers of inflammation, apoptosis, cardiac toxicity, oxidative stress, and upregulated anti-oxidative machinery. The histology of heart further supports our biochemical findings mitigated by the prophylactic treatment of myricetin. Henceforth, myricetin mitigates 5-FU-induced cardiac damage by modulating oxidative stress, inflammation, and cardiac-specific markers, as found in the present study.
Collapse
Affiliation(s)
- Azher Arafah
- Department
of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Muneeb U. Rehman
- Department
of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
- ,
| | - Ajaz Ahmad
- Department
of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Khalid M. AlKharfy
- Department
of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Saeed Alqahtani
- Department
of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Basit L. Jan
- Department
of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Nada M. Almatroudi
- Department
of Clinical Pharmacy, College of Pharmacy (Girls Campus), King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| |
Collapse
|
11
|
Abstract
Chemotherapy-induced gastrointestinal dysfunction is a common occurrence associated with many different classes of chemotherapeutic agents. Gastrointestinal toxicity includes mucositis, diarrhea, and constipation, and can often be a dose-limiting complication, induce cessation of treatment and could be life threatening. The gastrointestinal epithelium is rich in rapidly dividing cells and hence is a prime target for chemotherapeutic drugs. The incidence of gastrointestinal toxicity, including diarrhea and mucositis, is extremely high for a wide array of chemotherapeutic and radiation regimens. In fact, 60%-100% of patients on high-dose chemotherapy suffer from gastrointestinal side effects. Unfortunately, treatment options are limited, and therapy is often restricted to palliative care. Therefore, there is a great unmet therapeutic need for preventing and treating chemotherapy-induced gastrointestinal toxicities in the clinic. In this review, we discuss our current understanding of the mechanisms underlying chemotherapy-induced diarrhea and mucositis, and emerging mechanisms involving the enteric nervous system, smooth muscle cells and enteric immune cells. Recent evidence has also implicated gut dysbiosis in the pathogenesis of not only chemotherapy-induced mucositis and diarrhea, but also chemotherapy-induced peripheral neuropathy. Oxidative stress induced by chemotherapeutic agents results in post-translational modification of ion channels altering neuronal excitability. Thus, investigating how chemotherapy-induced changes in the gut- microbiome axis may lead to gut-related toxicities will be critical in the discovery of new drug targets for mitigating adverse gastrointestinal effects associated with chemotherapy treatment.
Collapse
Affiliation(s)
- Hamid I Akbarali
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States.
| | - Karan H Muchhala
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Donald K Jessup
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Stanley Cheatham
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
12
|
Dahlgren D, Sjöblom M, Hellström PM, Lennernäs H. Chemotherapeutics-Induced Intestinal Mucositis: Pathophysiology and Potential Treatment Strategies. Front Pharmacol 2021; 12:681417. [PMID: 34017262 PMCID: PMC8129190 DOI: 10.3389/fphar.2021.681417] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 04/19/2021] [Indexed: 12/11/2022] Open
Abstract
The gastrointestinal tract is particularly vulnerable to off-target effects of antineoplastic drugs because intestinal epithelial cells proliferate rapidly and have a complex immunological interaction with gut microbiota. As a result, up to 40-100% of all cancer patients dosed with chemotherapeutics experience gut toxicity, called chemotherapeutics-induced intestinal mucositis (CIM). The condition is associated with histological changes and inflammation in the mucosa arising from stem-cell apoptosis and disturbed cellular renewal and maturation processes. In turn, this results in various pathologies, including ulceration, pain, nausea, diarrhea, and bacterial translocation sepsis. In addition to reducing patient quality-of-life, CIM often leads to dose-reduction and subsequent decrease of anticancer effect. Despite decades of experimental and clinical investigations CIM remains an unsolved clinical issue, and there is a strong consensus that effective strategies are needed for preventing and treating CIM. Recent progress in the understanding of the molecular and functional pathology of CIM had provided many new potential targets and opportunities for treatment. This review presents an overview of the functions and physiology of the healthy intestinal barrier followed by a summary of the pathophysiological mechanisms involved in the development of CIM. Finally, we highlight some pharmacological and microbial interventions that have shown potential. Conclusively, one must accept that to date no single treatment has substantially transformed the clinical management of CIM. We therefore believe that the best chance for success is to use combination treatments. An optimal combination treatment will likely include prophylactics (e.g., antibiotics/probiotics) and drugs that impact the acute phase (e.g., anti-oxidants, apoptosis inhibitors, and anti-inflammatory agents) as well as the recovery phase (e.g., stimulation of proliferation and adaptation).
Collapse
Affiliation(s)
- David Dahlgren
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Markus Sjöblom
- Department of Neuroscience, Division of Physiology, Uppsala University, Uppsala, Sweden
| | - Per M Hellström
- Department of Medical Sciences, Gastroenterology/Hepatology, Uppsala University, Uppsala, Sweden
| | - Hans Lennernäs
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
13
|
Cavalcanti BC, Neto JBDA, Silva AADS, Barreto FS, Ferreira JRDO, Magalhães HIF, Silva CRD, Vieira ÍGP, Ricardo NMPS, Nobre Júnior HV, Moraes MO. Chemopreventive effect of troxerutin against hydrogen peroxide-induced oxidative stress in human leukocytes through modulation of glutathione-dependent enzymes. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2021; 84:137-151. [PMID: 33103637 DOI: 10.1080/15287394.2020.1836541] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Troxerutin is a natural flavonoid present abundantly in tea, coffee, olives, wheat, and a variety of fruits and vegetables. Due to its diverse pharmacological properties, this flavonoid has aroused interest for treatment of various diseases, and consequently prompted investigation into its toxicological characteristics. The aim of this study was to evaluate the genotoxic and mutagenic effects and chemoprotective activity attributed to troxerutin using human peripheral blood leukocytes (PBLs) through several well-established experimental protocols based upon different parameters. Data demonstrated that troxerutin (100 to 1000 µM) induced no marked cytotoxic effect on PBLs after 24 hr, and did not produce strand breaks and mutagenicity. Regarding chemoprevention, this flavonoid attenuated cytotoxicity, genotoxicity, and mutagenicity initiated by hydrogen peroxide (H2O2) in human PBLs. Further, troxerutin demonstrated no marked cytotoxic effect on PBLs and exerted a protective effect against oxidative stress induced by H2O2 through modulation of GSH-dependent enzymes.
Collapse
Affiliation(s)
- Bruno Coêlho Cavalcanti
- Drug Research and Development Center, Federal University of Ceará , Fortaleza, Brazil
- Department of Physiology and Pharmacology, Federal University of Ceará , Fortaleza, CE, Brazil
| | - João Batista de Andrade Neto
- Drug Research and Development Center, Federal University of Ceará , Fortaleza, Brazil
- School of Pharmacy, Laboratory for Bioprospection of Antimicrobial Molecules, Federal University of Ceará , Fortaleza, Brazil
- Christus University Center (UNICHRISTUS) , Fortaleza, Brazil
| | | | | | | | | | - Cecília Rocha da Silva
- Drug Research and Development Center, Federal University of Ceará , Fortaleza, Brazil
- School of Pharmacy, Laboratory for Bioprospection of Antimicrobial Molecules, Federal University of Ceará , Fortaleza, Brazil
| | | | | | - Hélio Vitoriano Nobre Júnior
- Drug Research and Development Center, Federal University of Ceará , Fortaleza, Brazil
- School of Pharmacy, Laboratory for Bioprospection of Antimicrobial Molecules, Federal University of Ceará , Fortaleza, Brazil
| | - Manoel Odorico Moraes
- Drug Research and Development Center, Federal University of Ceará , Fortaleza, Brazil
- Department of Physiology and Pharmacology, Federal University of Ceará , Fortaleza, CE, Brazil
| |
Collapse
|
14
|
Chen KJ, Chen YL, Ueng SH, Hwang TL, Kuo LM, Hsieh PW. Neutrophil elastase inhibitor (MPH-966) improves intestinal mucosal damage and gut microbiota in a mouse model of 5-fluorouracil-induced intestinal mucositis. Biomed Pharmacother 2021; 134:111152. [PMID: 33373916 DOI: 10.1016/j.biopha.2020.111152] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/03/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND 5-Fluorouracil (5-FU)-based chemotherapy is first-line chemotherapy for colorectal cancer. However, 5-FU-induced intestinal mucositis (FUIIM) is a common adverse effect that severely impairs drug tolerance and results in poor patient health. METHODS Male C57BL/6 mice were given 5-FU (50 mg/kg/day, i.p.) and treated with MPH-966 (5 and 7.5 mg/kg/day, p.o.) for five days. The body weight loss and the amount of food intake, and histopathological findings were recorded and analyzed. In addition, the neutrophil infiltration, levels of neutrophil serine proteases and pro-inflammatory cytokines, and tight junction proteins expression in intestinal tissues were determined. The ecology of gut microbiota was performed through next-generation sequencing technologies. RESULTS Neutrophil elastase (NE) overexpression is a key feature in FUIIM. This study showed that treatment with the specific NE inhibitor MPH-966 (7.5 mg/kg/day, p.o.) significantly reversed 5-FU-induced loss in body weight and food intake; reversed villous atrophy; significantly suppressed myeloperoxidase, NE, and proteinase 3 activity; and reduced pro-inflammatory cytokine expression in an FUIIM mouse model. In addition, MPH-966 prevented 5-FU-induced intestinal barrier dysfunction, as indicated by the modulated expression of the tight junction proteins zonula occludin-1 and occludin. MPH-966 also reversed 5-FU-induced changes in gut microbiota diversity and abundances, specifically the Firmicutes-to-Bacteroidetes ratio; Muribaculaceae, Ruminococcaceae, and Eggerthellaceae abundances at the family level; and Candidatus Arthromitus abundance at the genus level. CONCLUSION These data indicate that NE inhibitor is a key treatment candidate to alleviate FUIIM by regulating abnormal inflammatory responses, intestinal barrier dysfunction, and gut microbiota imbalance.
Collapse
Affiliation(s)
- Kung-Ju Chen
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Natural Products, School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Li Chen
- Graduate Institute of Natural Products, School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shir-Hwa Ueng
- Department of Pathology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Tsong-Long Hwang
- Graduate Institute of Natural Products, School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital, Linkou, Taiwan; Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan, Taiwan; Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan; Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City, Taiwan
| | - Liang-Mou Kuo
- Department of General Surgery, Chang Gung Memorial Hospital, Chiayi, 613, Taiwan.
| | - Pei-Wen Hsieh
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Natural Products, School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital, Linkou, Taiwan; Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan, Taiwan.
| |
Collapse
|
15
|
Li J, Zhang L, Li Y, Wu Y, Wu T, Feng H, Xu Z, Liu Y, Ruan Z, Zhou S. Puerarin improves intestinal barrier function through enhancing goblet cells and mucus barrier. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.104246] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
16
|
Fideles LDS, de Miranda JAL, Martins CDS, Barbosa MLL, Pimenta HB, Pimentel PVDS, Teixeira CS, Scafuri MAS, Façanha SDO, Barreto JEF, Carvalho PMDM, Scafuri AG, Araújo JL, Rocha JA, Vieira IGP, Ricardo NMPS, da Silva Campelo M, Ribeiro MENP, de Castro Brito GA, Cerqueira GS. Role of Rutin in 5-Fluorouracil-Induced Intestinal Mucositis: Prevention of Histological Damage and Reduction of Inflammation and Oxidative Stress. Molecules 2020; 25:molecules25122786. [PMID: 32560278 PMCID: PMC7356626 DOI: 10.3390/molecules25122786] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 06/12/2020] [Accepted: 06/14/2020] [Indexed: 02/07/2023] Open
Abstract
Intestinal mucositis, characterized by inflammatory and/or ulcerative processes in the gastrointestinal tract, occurs due to cellular and tissue damage following treatment with 5-fluorouracil (5-FU). Rutin (RUT), a natural flavonoid extracted from Dimorphandra gardneriana, exhibits antioxidant, anti-inflammatory, cytoprotective, and gastroprotective properties. However, the effect of RUT on inflammatory processes in the intestine, especially on mucositis promoted by antineoplastic agents, has not yet been reported. In this study, we investigated the role of RUT on 5-FU-induced experimental intestinal mucositis. Swiss mice were randomly divided into seven groups: Saline, 5-FU, RUT-50, RUT-100, RUT-200, Celecoxib (CLX), and CLX + RUT-200 groups. The mice were weighed daily. After treatment, the animals were euthanized and segments of the small intestine were collected to evaluate histopathological alterations (morphometric analysis); malondialdehyde (MDA), myeloperoxidase (MPO), and glutathione (GSH) concentrations; mast and goblet cell counts; and cyclooxygenase-2 (COX-2) activity, as well as to perform immunohistochemical analyses. RUT treatment (200 mg/kg) prevented 5-FU-induced histopathological changes and reduced oxidative stress by decreasing MDA concentrations and increasing GSH concentrations. RUT attenuated the inflammatory response by decreasing MPO activity, intestinal mastocytosis, and COX-2 expression. These results suggest that the COX-2 pathway is one of the underlying protective mechanisms of RUT against 5-FU-induced intestinal mucositis.
Collapse
Affiliation(s)
- Lázaro de Sousa Fideles
- Department of Morphology, Faculty of Medicine, Federal University of Ceará, s/n Delmiro of Farias Street, Porangabuçu Campus, Fortaleza 60416-030, Brazil; (L.d.S.F.); (C.d.S.M.); (M.L.L.B.); (H.B.P.); (P.V.d.S.P.); (C.S.T.); (J.E.F.B.); (A.G.S.); (G.A.d.C.B.); (G.S.C.)
| | - João Antônio Leal de Miranda
- Department of Morphology, Faculty of Medicine, Federal University of Ceará, s/n Delmiro of Farias Street, Porangabuçu Campus, Fortaleza 60416-030, Brazil; (L.d.S.F.); (C.d.S.M.); (M.L.L.B.); (H.B.P.); (P.V.d.S.P.); (C.S.T.); (J.E.F.B.); (A.G.S.); (G.A.d.C.B.); (G.S.C.)
- Correspondence: ; Tel.: +55-85-3366-8492
| | - Conceição da Silva Martins
- Department of Morphology, Faculty of Medicine, Federal University of Ceará, s/n Delmiro of Farias Street, Porangabuçu Campus, Fortaleza 60416-030, Brazil; (L.d.S.F.); (C.d.S.M.); (M.L.L.B.); (H.B.P.); (P.V.d.S.P.); (C.S.T.); (J.E.F.B.); (A.G.S.); (G.A.d.C.B.); (G.S.C.)
| | - Maria Lucianny Lima Barbosa
- Department of Morphology, Faculty of Medicine, Federal University of Ceará, s/n Delmiro of Farias Street, Porangabuçu Campus, Fortaleza 60416-030, Brazil; (L.d.S.F.); (C.d.S.M.); (M.L.L.B.); (H.B.P.); (P.V.d.S.P.); (C.S.T.); (J.E.F.B.); (A.G.S.); (G.A.d.C.B.); (G.S.C.)
| | - Helder Bindá Pimenta
- Department of Morphology, Faculty of Medicine, Federal University of Ceará, s/n Delmiro of Farias Street, Porangabuçu Campus, Fortaleza 60416-030, Brazil; (L.d.S.F.); (C.d.S.M.); (M.L.L.B.); (H.B.P.); (P.V.d.S.P.); (C.S.T.); (J.E.F.B.); (A.G.S.); (G.A.d.C.B.); (G.S.C.)
| | - Paulo Vitor de Souza Pimentel
- Department of Morphology, Faculty of Medicine, Federal University of Ceará, s/n Delmiro of Farias Street, Porangabuçu Campus, Fortaleza 60416-030, Brazil; (L.d.S.F.); (C.d.S.M.); (M.L.L.B.); (H.B.P.); (P.V.d.S.P.); (C.S.T.); (J.E.F.B.); (A.G.S.); (G.A.d.C.B.); (G.S.C.)
| | - Claudio Silva Teixeira
- Department of Morphology, Faculty of Medicine, Federal University of Ceará, s/n Delmiro of Farias Street, Porangabuçu Campus, Fortaleza 60416-030, Brazil; (L.d.S.F.); (C.d.S.M.); (M.L.L.B.); (H.B.P.); (P.V.d.S.P.); (C.S.T.); (J.E.F.B.); (A.G.S.); (G.A.d.C.B.); (G.S.C.)
| | | | | | - João Erivan Façanha Barreto
- Department of Morphology, Faculty of Medicine, Federal University of Ceará, s/n Delmiro of Farias Street, Porangabuçu Campus, Fortaleza 60416-030, Brazil; (L.d.S.F.); (C.d.S.M.); (M.L.L.B.); (H.B.P.); (P.V.d.S.P.); (C.S.T.); (J.E.F.B.); (A.G.S.); (G.A.d.C.B.); (G.S.C.)
- Christus University Center (Unichristus), 133 Adolfo Gurgel Street, Fortaleza 63010-475, Brazil;
| | | | - Ariel Gustavo Scafuri
- Department of Morphology, Faculty of Medicine, Federal University of Ceará, s/n Delmiro of Farias Street, Porangabuçu Campus, Fortaleza 60416-030, Brazil; (L.d.S.F.); (C.d.S.M.); (M.L.L.B.); (H.B.P.); (P.V.d.S.P.); (C.S.T.); (J.E.F.B.); (A.G.S.); (G.A.d.C.B.); (G.S.C.)
- Scafuri Institute of Human Sexuality, 1513 Republic of Lebanon Street, Varjota, Fortaleza 60175-222, Brazil;
| | - Joabe Lima Araújo
- Department of Genetics and Morphology, s/n Darcy Ribeiro University Campus, University of Brasília, Brasília-DF 70910-900, Brazil;
| | - Jefferson Almeida Rocha
- Medicinal Chemistry and Biotechnology Research Group (QUIMEBIO), Federal University of Maranhão (UFMA), São Bernardo/MA 65550-000, Brazil;
| | - Icaro Gusmão Pinto Vieira
- Technological Development Park, Federal University of Ceará, Humberto Monte Avenue, 2977, Pici Campus, Fortaleza 60440-900, Brazil;
| | - Nágila Maria Pontes Silva Ricardo
- Department of Organic and Inorganic Chemistry, Federal University of Ceará, Pici Campus, Fortaleza 60440-900, Brazil; (N.M.P.S.R.); (M.d.S.C.); (M.E.N.P.R.)
| | - Matheus da Silva Campelo
- Department of Organic and Inorganic Chemistry, Federal University of Ceará, Pici Campus, Fortaleza 60440-900, Brazil; (N.M.P.S.R.); (M.d.S.C.); (M.E.N.P.R.)
| | - Maria Elenir Nobre Pinho Ribeiro
- Department of Organic and Inorganic Chemistry, Federal University of Ceará, Pici Campus, Fortaleza 60440-900, Brazil; (N.M.P.S.R.); (M.d.S.C.); (M.E.N.P.R.)
| | - Gerly Anne de Castro Brito
- Department of Morphology, Faculty of Medicine, Federal University of Ceará, s/n Delmiro of Farias Street, Porangabuçu Campus, Fortaleza 60416-030, Brazil; (L.d.S.F.); (C.d.S.M.); (M.L.L.B.); (H.B.P.); (P.V.d.S.P.); (C.S.T.); (J.E.F.B.); (A.G.S.); (G.A.d.C.B.); (G.S.C.)
| | - Gilberto Santos Cerqueira
- Department of Morphology, Faculty of Medicine, Federal University of Ceará, s/n Delmiro of Farias Street, Porangabuçu Campus, Fortaleza 60416-030, Brazil; (L.d.S.F.); (C.d.S.M.); (M.L.L.B.); (H.B.P.); (P.V.d.S.P.); (C.S.T.); (J.E.F.B.); (A.G.S.); (G.A.d.C.B.); (G.S.C.)
| |
Collapse
|