1
|
Redeghieri P, Moray J, Kerff F, Gohy S, Leal T, Muyldermans S, Vanbever R, Morales‐Yánez FJ, Dumoulin M. Enzymatic, structural, and biophysical characterization of a single-domain antibody (VHH) selectively and tightly inhibiting neutrophil elastase and exhibiting favorable developability properties. Protein Sci 2024; 33:e5227. [PMID: 39604162 PMCID: PMC11602439 DOI: 10.1002/pro.5227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024]
Abstract
Human neutrophil elastase (hNE), a serine protease released by neutrophils during inflammation, plays a major role in the pathophysiology of several conditions especially in inflammatory lung diseases. Its inhibition constitutes, therefore, a promising therapeutic strategy to combat these diseases. In this work, we characterized the in vitro properties of a VHH (i.e., the antigen binding domain of camelid heavy chain-only antibodies), referred to as NbE201. This VHH is able to inhibit tightly, selectively and competitively both human and murine elastases with the inhibition constants (Ki) of 4.1 ± 0.9 nM and 36.8 ± 3.9 nM, respectively. The IC50 for the inhibition of the hydrolysis of elastin is in the same range to that of alpha-1 antitrypsin (i.e., the main endogenous inhibitor of hNE also used in the clinic) and 14 times better than that of Sivelestat (i.e., the 2nd clinically approved hNE inhibitor). The X-ray crystal structure of the NbE201-hNE complex reveals that the Complementarity Determining Regions CDR1 and CDR3 of the VHH bind into the substrate binding pocket of hNE and prevent the access to small or macromolecular substrates. They do not, however, bind deep enough into the pocket to be hydrolyzed. NbE201 is highly stable towards oxidation, deamidation, and chemical or thermal denaturation. NbE201 is therefore likely to tolerate manufacturing processes during drug development. These results highlight the high potential of NbE201 as a (pre)clinical tool to diagnose and treat diseases associated with excessive hNE activity, and for fundamental research to better understand the role of hNE in these conditions.
Collapse
Affiliation(s)
- Paola Redeghieri
- Nano‐Antibodies to Explore Protein Structure and Functions (NEPTUNS)Centre for Protein Engineering, InBios, Department of Life Sciences, University of LiègeLiègeBelgium
| | - Joël Moray
- Nano‐Antibodies to Explore Protein Structure and Functions (NEPTUNS)Centre for Protein Engineering, InBios, Department of Life Sciences, University of LiègeLiègeBelgium
| | - Frédéric Kerff
- Biological Macromolecule Crystallography, Centre for Protein Engineering, InBios, Department of Life SciencesUniversity of LiègeLiègeBelgium
| | - Sophie Gohy
- Department of Pneumology, ENT and Dermatology, Institute of Experimental and Clinical Research (IREC)Université Catholique de LouvainBrusselsBelgium
- Cystic Fibrosis Reference CentreCliniques Universitaires Saint‐LucBrusselsBelgium
| | - Teresinha Leal
- Louvain Center for Toxicology and Applied Pharmacology (LTAP), Institute of Experimental and Clinical Research (IREC)Université Catholique de LouvainBrusselsBelgium
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular ImmunologyVrije Universiteit BrusselBrusselsBelgium
| | - Rita Vanbever
- Louvain Drug Research Institute (LDRI)Université Catholique de LouvainBrusselsBelgium
| | - Francisco Javier Morales‐Yánez
- Nano‐Antibodies to Explore Protein Structure and Functions (NEPTUNS)Centre for Protein Engineering, InBios, Department of Life Sciences, University of LiègeLiègeBelgium
| | - Mireille Dumoulin
- Nano‐Antibodies to Explore Protein Structure and Functions (NEPTUNS)Centre for Protein Engineering, InBios, Department of Life Sciences, University of LiègeLiègeBelgium
| |
Collapse
|
2
|
Teng Y, Li J, Guo J, Yan C, Wang A, Xia X. Alginate oligosaccharide improves 5-fluorouracil-induced intestinal mucositis by enhancing intestinal barrier and modulating intestinal levels of butyrate and isovalerate. Int J Biol Macromol 2024; 276:133699. [PMID: 38972652 DOI: 10.1016/j.ijbiomac.2024.133699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 07/09/2024]
Abstract
Chemotherapy-induced mucositis (CIM) is the typical side effect of chemotherapy. This study investigates the potential of alginate oligosaccharide (AOS) in ameliorating CIM induced by 5-fluorouracil (5-FU) in a murine model and its underlying mechanisms. AOS effectively mitigated body weight loss and histopathological damage, modulated inflammatory cytokines and attenuated the oxidative stress. AOS restored intestinal barrier integrity through enhancing expression of tight junction proteins via MLCK signaling pathway. AOS alleviated intestinal mucosal damage by inhibiting TLR4/MyD88/NF-κB signaling pathway, downregulating the pro-apoptotic protein Bax and upregulating the anti-apoptotic protein Bcl-2. Moreover, AOS significantly enriched intestinal Akkermansiaceae and increased the production of short-chain fatty acids (SCFAs), most notably butyrate and isovalerate. Pre-treatment with butyrate and isovalerate also alleviated 5-FU-induced CIM. In conclusion, AOS effectively mitigated CIM through strenghthening intestinal barrier, attenuating inflammation, and modulating gut microbiota and intestianl levels of butyrate and isovalerate. These finding indicate that AOS could be potentially utilized as a supplemental strategy for prevention or mitigation of CIM.
Collapse
Affiliation(s)
- Yue Teng
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China; State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Jiahui Li
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China; State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Jian Guo
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China; State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Chunhong Yan
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China; State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Ailing Wang
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China; State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Xiaodong Xia
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China; State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China.
| |
Collapse
|
3
|
Wang JL, Yeh CH, Huang SH, Wu LSH, Chen MCM. Effects of Resistant-Starch-Encapsulated Probiotic Cocktail on Intestines Damaged by 5-Fluorouracil. Biomedicines 2024; 12:1912. [PMID: 39200376 PMCID: PMC11351836 DOI: 10.3390/biomedicines12081912] [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: 06/20/2024] [Revised: 08/06/2024] [Accepted: 08/16/2024] [Indexed: 09/02/2024] Open
Abstract
Probiotics and prebiotics have gained attention for their potential health benefits. However, their efficacy hinges on probiotic survival through the harsh gastrointestinal environment. Microencapsulation techniques provide a solution, with resistant starch (RS)-based techniques showing promise in maintaining probiotic viability. Specifically, RS-encapsulated probiotics significantly improved probiotic survival in gastric acid, bile salts, and simulated intestinal conditions. This study investigated the effects of a resistant-starch-encapsulated probiotic cocktail (RS-Pro) in the context of 5-fluorouracil (5-FU) chemotherapy, which frequently induces microbiota dysbiosis and intestinal mucositis. Female BALB/c mice were divided into three groups: a 5-FU group, a 5-FU+Pro group receiving free probiotics, and a 5-FU+RS-Pro group receiving RS-encapsulated probiotics. After 28 days of treatment, analyses were conducted on fecal microbiota, intestinal histology, peripheral blood cell counts, and body and organ weights. It was revealed by 16S rRNA MiSeq sequencing that 5-FU treatment disrupted gut microbiota composition, reduced microbial diversity, and caused dysbiosis. RS-Pro treatment restored microbial diversity and increased the population of beneficial bacteria, such as Muribaculaceae, which play roles in carbohydrate and polyphenol metabolism. Furthermore, 5-FU administration induced moderate intestinal mucositis, characterized by reduced cellularity and shortened villi. However, RS-Pro treatment attenuated 5-FU-induced intestinal damage, preserving villus length. Mild leukopenia observed in the 5-FU-treated mice was partially alleviated in 5-FU+Pro and 5-FU+RS-Pro groups. These findings suggest that RS-Pro may serve as an adjunct to chemotherapy, potentially reducing adverse effects and improving therapeutic outcomes in future clinical applications.
Collapse
Affiliation(s)
- Jui-Ling Wang
- Animal Testing Division, National Applied Research Laboratories, National Laboratory Animal Center, Tainan 744, Taiwan;
| | - Chin-Hsing Yeh
- Fecula Biotech Co., Ltd., Tainan 744, Taiwan; (C.-H.Y.); (S.-H.H.)
| | - Shih-Hung Huang
- Fecula Biotech Co., Ltd., Tainan 744, Taiwan; (C.-H.Y.); (S.-H.H.)
| | - Lawrence Shih-Hsin Wu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404, Taiwan
| | | |
Collapse
|
4
|
Wang XH, Dong PF, Wang F, Zhou L. Schisandrin A can promote the anti-tumor effect of 5-Fu by reversing the immunosuppressive state of the body in rat. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2024:1-18. [PMID: 38979968 DOI: 10.1080/10286020.2024.2375291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 06/28/2024] [Indexed: 07/10/2024]
Abstract
In this study, based on Walker 256 in vitro experiments, CCK-8 assay, clone formation assay, wound healing assay, and flow cytometry were used to detect cell apoptosis and cell cycle. It was found that schisandrin may have significant anti-tumor effects in vitro by inhibiting TGF-β/Smad signaling pathway. In addition, in vivo experiments, immunohistochemistry was used to observe the expression of HIF-1α, VEGF and VEGFR-2 in tumor tissues. It was found that schisandrin could significantly improve the immunosuppression induced by 5-Fu and enhance the antitumor effect of 5-Fu. The mechanism may be related to the inhibition of Wnt-1/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Xiao-Hui Wang
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China
- Department of Interventional Therapy, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Peng-Fei Dong
- Department of Traditional Chinese Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450001, China
| | - Feng Wang
- Department of Interventional Therapy, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Lin Zhou
- Department of Pharmacology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
5
|
Aquilani R, Brugnatelli S, Maestri R, Iadarola P, Corallo S, Pagani A, Serra F, Bellini A, Buonocore D, Dossena M, Boschi F, Verri M. Chemotherapy-Induced Changes in Plasma Amino Acids and Lipid Oxidation of Resected Patients with Colorectal Cancer: A Background for Future Studies. Int J Mol Sci 2024; 25:5300. [PMID: 38791339 PMCID: PMC11121634 DOI: 10.3390/ijms25105300] [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: 04/13/2024] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Previous studies have documented that FOLFOX and XELOX therapies negatively impact the metabolism of skeletal muscle and extra-muscle districts. This pilot study tested whether three-month FOLFOX or XELOX therapy produced changes in plasma amino acid levels (PAAL) (an estimation of whole-body amino acid metabolism) and in plasma levels of malondialdehyde (MDA), a marker of lipid hyper oxidation. Fourteen ambulatory, resected patients with colorectal cancer scheduled to receive FOLFOX (n = 9) or XELOX (n = 5) therapy, after overnight fasting, underwent peripheral venous blood sampling, to determine PAAL and MDA before, during, and at the end of three-month therapy. Fifteen healthy matched subjects (controls) only underwent measures of PAAL at baseline. The results showed changes in 87.5% of plasma essential amino acids (EAAs) and 38.4% of non-EAAs in patients treated with FOLFOX or XELOX. These changes in EAAs occurred in two opposite directions: EAAs decreased with FOLFOX and increased or did not decrease with XELOX (interactions: from p = 0.034 to p = 0.003). Baseline plasma MDA levels in both FOLFOX and XELOX patients were above the normal range of values, and increased, albeit not significantly, during therapy. In conclusion, three-month FOLFOX or XELOX therapy affected plasma EAAs differently but not the baseline MDA levels, which were already high.
Collapse
Affiliation(s)
- Roberto Aquilani
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, 27100 Pavia, Italy; (R.A.); (P.I.); (A.B.); (D.B.); (M.D.)
| | - Silvia Brugnatelli
- Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (S.B.); (S.C.); (A.P.); (F.S.)
| | - Roberto Maestri
- Department of Biomedical Engineering of the Montescano Institute, Istituti Clinici Scientifici Maugeri IRCCS, 27040 Montescano, Italy;
| | - Paolo Iadarola
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, 27100 Pavia, Italy; (R.A.); (P.I.); (A.B.); (D.B.); (M.D.)
| | - Salvatore Corallo
- Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (S.B.); (S.C.); (A.P.); (F.S.)
| | - Anna Pagani
- Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (S.B.); (S.C.); (A.P.); (F.S.)
| | - Francesco Serra
- Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (S.B.); (S.C.); (A.P.); (F.S.)
| | - Anna Bellini
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, 27100 Pavia, Italy; (R.A.); (P.I.); (A.B.); (D.B.); (M.D.)
| | - Daniela Buonocore
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, 27100 Pavia, Italy; (R.A.); (P.I.); (A.B.); (D.B.); (M.D.)
| | - Maurizia Dossena
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, 27100 Pavia, Italy; (R.A.); (P.I.); (A.B.); (D.B.); (M.D.)
| | - Federica Boschi
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy;
| | - Manuela Verri
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, 27100 Pavia, Italy; (R.A.); (P.I.); (A.B.); (D.B.); (M.D.)
| |
Collapse
|
6
|
Shin HJ, Yang WK, Lee YC, Kim S, Moon SO, Kwon YJ, Noh HJ, Kim KH, Kim BK, Shin CH, Chae MY, Yun SH, Kim SH. Protective effect of the mixture of Lactiplantibacillus plantarum KC3 and Leonurus Japonicas Houtt extract on respiratory disorders. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 270:115856. [PMID: 38134637 DOI: 10.1016/j.ecoenv.2023.115856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/07/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023]
Abstract
Air pollutants, such as particulate matter (PM) and diesel exhaust particles (DEP), are associated with respiratory diseases. Therefore, preventive and therapeutic strategies against PM-and DEP (PM10D)-induced respiratory diseases are needed. Herein, we evaluate the protective effects of a mixture of Lactiplantibacillus plantarum KC3 and Leonurus Japonicas Houtt (LJH) extract against airway inflammation associated with exposure to PM10D. To determine the anti-inflammatory effects of the LJH extract, reactive oxygen species (ROS) production and the expression of inflammatory pathways were determined in PM10-induced MH-S cells. For the respiratory protective effects, BALB/c mice were exposed to PM10D via intranasal injection, and a mixture of L. plantarum KC3 and LJH extract was administered orally for 12 days. LJH extract inhibited ROS production and the phosphorylation of downstream factors of NF-κB in PM10-stimulated MH-S cells. The mixture of L. plantarum KC3 and LJH repressed the infiltration of neutrophils, reduced the immune cells number, and suppressed the proinflammatory mediators and cyclooxygenase (COX)-2 expressions in PM10D-induced airway inflammation with reduced phosphorylation of downstream factors of NF-κB. In addition, these effects were not observed in an alveolar macrophage depleted PM10D-induced mouse model using clodronate liposomes. The extract mixture also regulated gut microbiota in feces and upregulated the mRNA expression of Foxp3, transforming growth factor (TGF)-β1, and interleukin (IL)-10 in the colon. The L. plantarum KC3 and LJH extract mixture may inhibit alveolar macrophage- and neutrophil-mediated inflammatory responses and regulate gut microbiota and immune response in PM10D-induced airway inflammation, suggesting it is a potential remedy to prevent and cure airway inflammation and respiratory disorders.
Collapse
Affiliation(s)
- Han Jae Shin
- KT&G Research Institute, Daejeon 34128, the Republic of Korea
| | - Won-Kyung Yang
- Institute of Traditional Medicine and Bioscience, Daejeon University, Daejeon 34520, the Republic of Korea
| | - Young Chul Lee
- KT&G Research Institute, Daejeon 34128, the Republic of Korea
| | - Soeun Kim
- KT&G Research Institute, Daejeon 34128, the Republic of Korea
| | - Sung Ok Moon
- KT&G Research Institute, Daejeon 34128, the Republic of Korea
| | - Yoo Jin Kwon
- Chong Kun Dang Bio Research Institute (CKDBiO), Seoul 03722, the Republic of Korea
| | - Hye-Ji Noh
- Chong Kun Dang Bio Research Institute (CKDBiO), Seoul 03722, the Republic of Korea
| | - Kyung Hwan Kim
- Chong Kun Dang Bio Research Institute (CKDBiO), Seoul 03722, the Republic of Korea
| | - Byoung Kook Kim
- Chong Kun Dang Bio Research Institute (CKDBiO), Seoul 03722, the Republic of Korea
| | - Chang Hun Shin
- Chong Kun Dang Bio Research Institute (CKDBiO), Seoul 03722, the Republic of Korea
| | - Min-Young Chae
- Institute of Traditional Medicine and Bioscience, Daejeon University, Daejeon 34520, the Republic of Korea
| | - Su-Hyeon Yun
- Institute of Traditional Medicine and Bioscience, Daejeon University, Daejeon 34520, the Republic of Korea
| | - Seung-Hyung Kim
- Institute of Traditional Medicine and Bioscience, Daejeon University, Daejeon 34520, the Republic of Korea.
| |
Collapse
|
7
|
Hwang TL, Lin JY, Kuo LM, Kumar Dhandabani G, Hsieh PW. Design and synthesis of sirtinol analogs as human neutrophil elastase inhibitors. Bioorg Med Chem Lett 2024; 97:129544. [PMID: 37939864 DOI: 10.1016/j.bmcl.2023.129544] [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: 07/21/2023] [Revised: 09/22/2023] [Accepted: 11/04/2023] [Indexed: 11/10/2023]
Abstract
Human neutrophil elastase (HNE) overexpression has a crucial role in most acute inflammation and alpha1-antitrypsin deficiency syndromes observed in humans, triggering neutrophil invasion and activation of macrophage inflammatory and proteolytic effects, leading to tissue damage. Manipulating HNE level homeostasis could potentially help treat neutrophilic inflammation. Previous studies have shown that sirtinol (1) has a specific influence on HNE and potently attenuates acute lung injury and hepatic injury mediated by lipopolysaccharide or trauma hemorrhage. Therefore, 1 was chosen as the model structure to obtain more potent anti-HNE agents. In the present study, we synthesized a series of sirtinol analogues and determined their inhibitory effects on HNE. Structure-activity relationship (SAR) studies showed that swapping the imine and methyl groups of the sirtinol scaffold with diazene and carboxyl groups, respectively, enhances the HNE inhibiting potency. Compound 29 exhibited the highest potency in the SAR study and showed dual inhibitory effects on HNE and proteinase 3 with IC50 values of 4.91 and 20.69 µM, respectively. Furthermore, 29 was confirmed to have dual impacts on inhibiting O2•- generation and elastase release in cell-based assays with IC50 values of 0.90 and 1.86 µM, respectively. These findings suggest that 29 is a promising candidate for developing HNE inhibitors in the treatment of neutrophilic inflammatory diseases.
Collapse
Affiliation(s)
- Tsong-Long Hwang
- Graduate Institute of Natural Products, School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Biomedical Sciences, 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.
| | - Jing-Yi Lin
- Graduate Institute of Natural Products, School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Liang-Mou Kuo
- Department of General Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan.
| | - Ganesh Kumar Dhandabani
- Graduate Institute of Natural Products, School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Pei-Wen Hsieh
- Graduate Institute of Natural Products, School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan, Taiwan; Department of General Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan.
| |
Collapse
|
8
|
Bowen J, Cross C. The Role of the Innate Immune Response in Oral Mucositis Pathogenesis. Int J Mol Sci 2023; 24:16314. [PMID: 38003503 PMCID: PMC10670995 DOI: 10.3390/ijms242216314] [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: 10/07/2023] [Revised: 11/11/2023] [Accepted: 11/12/2023] [Indexed: 11/26/2023] Open
Abstract
Oral mucositis (OM) is a significant complication of cancer therapy with limited management strategies. Whilst inflammation is a central feature of destructive and ultimately ulcerative pathology, to date, attempts to mitigate damage via this mechanism have proven limited. A relatively underexamined aspect of OM development is the contribution of elements of the innate immune system. In particular, the role played by barriers, pattern recognition systems, and microbial composition in early damage signaling requires further investigation. As such, this review highlights the innate immune response as a potential focus for research to better understand OM pathogenesis and development of interventions for patients treated with radiotherapy and chemotherapy. Future areas of evaluation include manipulation of microbial-mucosal interactions to alter cytotoxic sensitivity, use of germ-free models, and translation of innate immune-targeted agents interrogated for mucosal injury in other regions of the alimentary canal into OM-based clinical trials.
Collapse
Affiliation(s)
- Joanne Bowen
- School of Biomedicine, University of Adelaide, Adelaide 5005, Australia;
| | | |
Collapse
|
9
|
Huang B, Gui M, An H, Shen J, Ye F, Ni Z, Zhan H, Che L, Lai Z, Zeng J, Peng J, Lin J. Babao Dan alleviates gut immune and microbiota disorders while impacting the TLR4/MyD88/NF-кB pathway to attenuate 5-Fluorouracil-induced intestinal injury. Biomed Pharmacother 2023; 166:115387. [PMID: 37643486 DOI: 10.1016/j.biopha.2023.115387] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 08/31/2023] Open
Abstract
Adjuvant chemotherapy based on 5-fluorouracil (5-FU), such as FOLFOX, is suggested as a treatment for gastrointestinal cancer. Yet, intestinal damage continues to be a prevalent side effect for which there are no practical prevention measures. We investigated whether Babao Dan (BBD), a Traditional Chinese Medicine, protects against intestinal damage induced by 5-FU by controlling immune response and gut microbiota. 5-FU was injected intraperitoneally to establish the mice model, then 250 mg/kg BBD was gavaged for five days straight. 5-FU led to marked weight loss, diarrhea, fecal blood, and histopathologic intestinal damage. Administration of BBD reduced these symptoms, inhibited proinflammatory cytokine (IL-6, IL-1β, IFN-γ, TNF-α) secretion, and upregulated the ratio of CD3(+) T cells and the CD4(+)/CD8(+) ratio. According to 16S rRNA sequencing, BBD dramatically repaired the disruption of the gut microbiota caused in a time-dependent way, and increased the Firmicutes/Bacteroidetes (F/B) ratio. Transcriptomic results showed that the mechanism is mainly concentrated on the NF-κB pathway, and we found that BBD reduced the concentration of LPS in the fecal suspension and serum, and inhibited TLR4/MyD88/NF-κB pathway activation. Furthermore, at the genus level on the fifth day, BBD upregulated the abundance of unidentified_Corynebacteriaceae, Aerococcus, Blautia, Jeotgalicoccus, Odoribacter, Roseburia, Rikenella, Intestinimonas, unidentified_Lachnospiraceae, Enterorhabdus, Ruminiclostridium, and downregulated the abundance of Bacteroides, Parabacteroides, Parasutterella, Erysipelatoclostridium, which were highly correlated with intestinal injury or the TLR4/MyD88/NF-κB pathway. In conclusion, we established a network involving 5-FU, BBD, the immune response, gut microbiota, and key pathways to explain the pharmacology of oral BBD in preventing 5-FU-induced intestinal injury.
Collapse
Affiliation(s)
- Bin Huang
- Academy of Integrative Medicine of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, PR China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, PR China; Key Laboratory of Integrative Medicine of Fujian Province University, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, PR China
| | - Mengxuan Gui
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, PR China
| | - Honglin An
- Key Laboratory of Integrative Medicine of Fujian Province University, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, PR China
| | - Jiayu Shen
- Key Laboratory of Integrative Medicine of Fujian Province University, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, PR China
| | - Feimin Ye
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, PR China
| | - Zhuona Ni
- Academy of Integrative Medicine of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, PR China
| | - Hanzhang Zhan
- Key Laboratory of Integrative Medicine of Fujian Province University, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, PR China
| | - Li Che
- Xiamen Traditional Chinese Medicine Co., Ltd., Xiamen 361100, PR China
| | - Zhicheng Lai
- Xiamen Traditional Chinese Medicine Co., Ltd., Xiamen 361100, PR China
| | - Jiahan Zeng
- Xiamen Traditional Chinese Medicine Co., Ltd., Xiamen 361100, PR China
| | - Jun Peng
- Academy of Integrative Medicine of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, PR China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, PR China; Key Laboratory of Integrative Medicine of Fujian Province University, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, PR China
| | - Jiumao Lin
- Academy of Integrative Medicine of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, PR China; Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, PR China; Key Laboratory of Integrative Medicine of Fujian Province University, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, PR China.
| |
Collapse
|
10
|
Xia J, Dai QL, He S, Jia HJ, Liu XG, Hua H, Zhou M, Wang X. Artesunate alleviates 5-fluorouracil-induced intestinal damage by suppressing cellular senescence and enhances its antitumor activity. Discov Oncol 2023; 14:139. [PMID: 37498338 PMCID: PMC10374509 DOI: 10.1007/s12672-023-00747-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 07/06/2023] [Indexed: 07/28/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the most prevalent diagnosed malignancies and one of the leading causes of cancer-related deaths worldwide. 5-Fluorouracil (5-FU) and its combination regimen are commonly used as primary chemotherapeutic agents for advanced CRC. Intestinal mucositis is one of the most frequent side effects of 5-FU. Artesunate (Arte) is derived from the wormwood plant Artemisia annua. Arte is not only effective against malaria but also diabetes, atherosclerosis, inflammation, and other conditions. The mechanism by which 5-FU damages the intestinal tract is unclear, and there is no standard treatment for diarrhea caused by 5-FU. Therefore, it is critical to discover novel and promising therapeutic drugs for 5-FU side effect treatment. METHODS The morphology and expression of genes and proteins associated with the aging of HUVECs, HIECs, and intestinal tissues were compared to the those of the control group. The cell lines and tissues were evaluated by SA-β-Gal staining, Western blotting, and RT‒qPCR. HIEC and HCT116 cell viability was assessed in vitro by a CCK-8 assay and in vivo by a subcutaneous tumor mouse assay. Tumor cell proliferation and apoptosis was evaluated by immunohistochemistry. RESULTS Here, we report that Arte alleviates the adverse side effects caused by 5-FU in intestinal tissue, and that 5-FU-induced intestinal damage is associated with drug-induced chemical inflammation and an increase in the proportion of senescent cells. Arte decreases the ratio of SA-β-Gal-positive cells and downregulated the expression of aging-related proteins (p53, p16) and aging-related genes (p53, p21). Mechanistically, Arte relieves intestinal injury by inhibiting mTOR expression, which is associated with the regulation of aging. Moreover, Arte suppresses the p38MAPK and NF-κB signaling pathways, which are related to inflammation regulation. In addition, the combined therapy of Arte plus 5-FU significantly decreases cancer cell viability in vitro. Arte and 5-FU synergistically reduce the growth of colorectal cancer (CRC) xenografts in vivo. CONCLUSIONS Overall, our findings point to the crucial treatment effect of Arte on inflammation, intestinal cell senescence, and CRC cell proliferation and offer a new option for CRC treatment.
Collapse
Affiliation(s)
- Jing Xia
- School of Basic Medicine, Dali University, Dali, 671000, Yunnan, China
- Key Laboratory of University Cell Biology Yunnan Province, Dali, 671000, Yunnan, China
| | - Qian Long Dai
- School of Basic Medicine, Dali University, Dali, 671000, Yunnan, China
- Key Laboratory of University Cell Biology Yunnan Province, Dali, 671000, Yunnan, China
| | - Siyue He
- School of Basic Medicine, Dali University, Dali, 671000, Yunnan, China
- Key Laboratory of University Cell Biology Yunnan Province, Dali, 671000, Yunnan, China
| | - Hui-Jie Jia
- School of Basic Medicine, Dali University, Dali, 671000, Yunnan, China
- Key Laboratory of University Cell Biology Yunnan Province, Dali, 671000, Yunnan, China
| | - Xian-Guo Liu
- Department of Oncology, The Affiliated Chengdu 363 Hospital of Southwest Medical University, No. 108, Daosangshu Street, Chengdu, 610041, China
| | - Hui Hua
- School of Basic Medicine, Dali University, Dali, 671000, Yunnan, China
- Key Laboratory of University Cell Biology Yunnan Province, Dali, 671000, Yunnan, China
| | - Min Zhou
- School of Basic Medicine, Dali University, Dali, 671000, Yunnan, China.
- Key Laboratory of University Cell Biology Yunnan Province, Dali, 671000, Yunnan, China.
| | - Xiaobo Wang
- School of Basic Medicine, Dali University, Dali, 671000, Yunnan, China.
- Key Laboratory of University Cell Biology Yunnan Province, Dali, 671000, Yunnan, China.
| |
Collapse
|
11
|
Chen Z, Dong Y, Yan Q, Li Q, Yu C, Lai Y, Tan J, Fan M, Xu C, Li L, Shen W, Gu J, Cheng H, Sun D. Liquid chromatography-tandem mass spectrometry analysis of a ratio-optimized drug pair of Sophora flavescens Aiton and Coptis chinensis Franch and study on the mechanism of anti-colorectal cancer effect of two alkaloids thereof. Front Oncol 2023; 13:1198467. [PMID: 37404762 PMCID: PMC10316516 DOI: 10.3389/fonc.2023.1198467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 05/15/2023] [Indexed: 07/06/2023] Open
Abstract
The drug pair consisting of Sophora flavescens Aiton (Sophorae flavescentis radix, Kushen) and Coptis chinensis Franch. (Coptidis rhizoma, Huanglian), as described in Prescriptions for Universal Relief (Pujifang), is widely used to treat laxation. Matrine and berberine are the major active components of Kushen and Huanglian, respectively. These agents have shown remarkable anti-cancer and anti-inflammatory effects. A mouse model of colorectal cancer was used to determine the most effective combination of Kushen and Huanglian against anti-colorectal cancer. The results showed that the combination of Kushen and Huanglian at a 1:1 ratio exerted the best anti-colorectal cancer effect versus other ratios. Moreover, the anti-colorectal cancer effect and potential mechanism underlying the effects of matrine and berberine were evaluated by the analysis of combination treatment or monotherapy. In addition, the chemical constituents of Kushen and Huanglian were identified and quantified by liquid chromatography-tandem mass spectrometry (LC-MS/MS). A total of 67 chemical components were identified from the Kushen-Huanglian drug pair (water extraction), and the levels of matrine and berberine were 129 and 232 µg/g, respectively. Matrine and berberine reduced the growth of colorectal cancer and relieved the pathological conditions in mice. In addition, the combination of matrine and berberine displayed better anti-colorectal cancer efficacy than monotherapy. Moreover, matrine and berberine reduced the relative abundance of Bacteroidota and Campilobacterota at phylum level and that of Helicobacter, Lachnospiraceae_NK4A136_group, Candidatus_Arthromitus, norank_f_Lachnospiraceae, Rikenella, Odoribacter, Streptococcus, norank_f_Ruminococcaceae, and Anaerotruncus at the genus level. Western blotting results demonstrated that treatment with matrine and berberine decreased the protein expressions of c-MYC and RAS, whereas it increased that of sirtuin 3 (Sirt3). The findings indicated that the combination of matrine and berberine was more effective in inhibiting colorectal cancer than monotherapy. This beneficial effect might depend on the improvement of intestinal microbiota structure and regulation of the RAS/MEK/ERK-c-MYC-Sirt3 signaling axis.
Collapse
Affiliation(s)
- Zihan Chen
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yingying Dong
- Jiangsu Collaborative Innovation Center of Traditional Chinese Prevention and Treatment of Tumor Research Center for Theory and Application of Cancer Toxin Pathogenesis, Nanjing, China
| | - Qiuying Yan
- Jiangsu Collaborative Innovation Center of Traditional Chinese Prevention and Treatment of Tumor Research Center for Theory and Application of Cancer Toxin Pathogenesis, Nanjing, China
| | - Qin Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chengtao Yu
- Jiangsu Collaborative Innovation Center of Traditional Chinese Prevention and Treatment of Tumor Research Center for Theory and Application of Cancer Toxin Pathogenesis, Nanjing, China
| | - Yueyang Lai
- Jiangsu Collaborative Innovation Center of Traditional Chinese Prevention and Treatment of Tumor Research Center for Theory and Application of Cancer Toxin Pathogenesis, Nanjing, China
| | - Jiani Tan
- Jiangsu Collaborative Innovation Center of Traditional Chinese Prevention and Treatment of Tumor Research Center for Theory and Application of Cancer Toxin Pathogenesis, Nanjing, China
| | - Minmin Fan
- Jiangsu Collaborative Innovation Center of Traditional Chinese Prevention and Treatment of Tumor Research Center for Theory and Application of Cancer Toxin Pathogenesis, Nanjing, China
| | - Changliang Xu
- Jiangsu Collaborative Innovation Center of Traditional Chinese Prevention and Treatment of Tumor Research Center for Theory and Application of Cancer Toxin Pathogenesis, Nanjing, China
| | - Liu Li
- Jiangsu Collaborative Innovation Center of Traditional Chinese Prevention and Treatment of Tumor Research Center for Theory and Application of Cancer Toxin Pathogenesis, Nanjing, China
| | - Weixing Shen
- Jiangsu Collaborative Innovation Center of Traditional Chinese Prevention and Treatment of Tumor Research Center for Theory and Application of Cancer Toxin Pathogenesis, Nanjing, China
| | - Junfei Gu
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Haibo Cheng
- Jiangsu Collaborative Innovation Center of Traditional Chinese Prevention and Treatment of Tumor Research Center for Theory and Application of Cancer Toxin Pathogenesis, Nanjing, China
| | - Dongdong Sun
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Prevention and Treatment of Tumor Research Center for Theory and Application of Cancer Toxin Pathogenesis, Nanjing, China
| |
Collapse
|
12
|
Huang J, Hwang AYM, Jia Y, Kim B, Iskandar M, Mohammed AI, Cirillo N. Experimental Chemotherapy-Induced Mucositis: A Scoping Review Guiding the Design of Suitable Preclinical Models. Int J Mol Sci 2022; 23:15434. [PMID: 36499758 PMCID: PMC9737148 DOI: 10.3390/ijms232315434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/01/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022] Open
Abstract
Mucositis is a common and most debilitating complication associated with the cytotoxicity of chemotherapy. The condition affects the entire alimentary canal from the mouth to the anus and has a significant clinical and economic impact. Although oral and intestinal mucositis can occur concurrently in the same individual, these conditions are often studied independently using organ-specific models that do not mimic human disease. Hence, the purpose of this scoping review was to provide a comprehensive yet systematic overview of the animal models that are utilised in the study of chemotherapy-induced mucositis. A search of PubMed/MEDLINE and Scopus databases was conducted to identify all relevant studies. Multiple phases of filtering were conducted, including deduplication, title/abstract screening, full-text screening, and data extraction. Studies were reported according to the updated Preferred Reporting Items for Systematic reviews and Meta-Analyses Extension for Scoping Reviews (PRISMA-ScR) guidelines. An inter-rater reliability test was conducted using Cohen's Kappa score. After title, abstract, and full-text screening, 251 articles met the inclusion criteria. Seven articles investigated both chemotherapy-induced intestinal and oral mucositis, 198 articles investigated chemotherapy-induced intestinal mucositis, and 46 studies investigated chemotherapy-induced oral mucositis. Among a total of 205 articles on chemotherapy-induced intestinal mucositis, 103 utilised 5-fluorouracil, 34 irinotecan, 16 platinum-based drugs, 33 methotrexate, and 32 other chemotherapeutic agents. Thirteen articles reported the use of a combination of 5-fluorouracil, irinotecan, platinum-based drugs, or methotrexate to induce intestinal mucositis. Among a total of 53 articles on chemotherapy-induced oral mucositis, 50 utilised 5-fluorouracil, 2 irinotecan, 2 methotrexate, 1 topotecan and 1 with other chemotherapeutic drugs. Three articles used a combination of these drugs to induce oral mucositis. Various animal models such as mice, rats, hamsters, piglets, rabbits, and zebrafish were used. The chemotherapeutic agents were introduced at various dosages via three routes of administration. Animals were mainly mice and rats. Unlike intestinal mucositis, most oral mucositis models combined mechanical or chemical irritation with chemotherapy. In conclusion, this extensive assessment of the literature revealed that there was a large variation among studies that reproduce oral and intestinal mucositis in animals. To assist with the design of a suitable preclinical model of chemotherapy-induced alimentary tract mucositis, animal types, routes of administration, dosages, and types of drugs were reported in this study. Further research is required to define an optimal protocol that improves the translatability of findings to humans.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Nicola Cirillo
- Melbourne Dental School, The University of Melbourne, Carlton, VIC 3053, Australia
| |
Collapse
|
13
|
Interaction of microbiome and immunity in tumorigenesis and clinical treatment. Biomed Pharmacother 2022; 156:113894. [DOI: 10.1016/j.biopha.2022.113894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/12/2022] [Accepted: 10/17/2022] [Indexed: 11/15/2022] Open
|
14
|
Gao X, Hu Y, Tao Y, Liu S, Chen H, Li J, Zhao Y, Sheng J, Tian Y, Fan Y. Cymbopogon citratus (DC.) Stapf aqueous extract ameliorates loperamide-induced constipation in mice by promoting gastrointestinal motility and regulating the gut microbiota. Front Microbiol 2022; 13:1017804. [PMID: 36267178 PMCID: PMC9578511 DOI: 10.3389/fmicb.2022.1017804] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/09/2022] [Indexed: 12/02/2022] Open
Abstract
Slow transit constipation (STC) is the most common type of functional constipation. Drugs with good effects and few side effects are urgently needed form the treatment of STC. Cymbopogon citratus (DC.) Stapf (CC) is an important medicinal and edible spice plant. The wide range of biological activities suggested that CC may have laxative effects, but thus far, it has not been reported. In this study, the loperamide-induced STC mouse model was used to evaluate the laxative effect of the aqueous extract of CC (CCAE), and the laxative mechanism was systematically explored from the perspectives of the enteric nervous system (ENS), neurotransmitter secretion, gastrointestinal motility factors, intestinal inflammation, gut barrier and gut microbiota. The results showed that CCAE not only decreased the serum vasoactive intestinal polypeptide (VIP), induced nitric oxide synthases (iNOS), and acetylcholinesterase (AchE) in STC mice but also increased the expression of gastrointestinal motility factors in colonic interstitial cells of Cajal (ICCs) and smooth muscle cells (SMCs), thereby significantly shortening the defecation time and improving the gastrointestinal transit rate. The significantly affected gastrointestinal motility factors included stem cell factor receptor (c-Kit), stem cell factor (SCF), anoctamin 1 (Ano1), ryanodine receptor 3 (RyR3), smooth muscle myosin light chain kinase (smMLCK) and Connexin 43 (Cx43). Meanwhile, CCAE could repair loperamide-induced intestinal inflammation and intestinal barrier damage by reducing the expression of the pro-inflammatory factor IL-1β and increasing the expression of the anti-inflammatory factor IL-10, chemical barrier (Muc-2) and mechanical barrier (Cldn4, Cldn12, Occludin, ZO-1, and ZO-2). Interestingly, CCAE could also partially restore loperamide-induced gut microbial dysbiosis in various aspects, such as microbial diversity, community structure and species composition. Importantly, we established a complex but clear network between gut microbiota and host parameters. Muribaculaceae, Lachnospiraceae and UCG-010 showed the most interesting associations with the laxative phenotypes; several other specific taxa showed significant associations with serum neurotransmitters, gastrointestinal motility factors, intestinal inflammation, and the gut barrier. These findings suggested that CCAE might promote intestinal motility by modulating the ENS-ICCs-SMCs network, intestinal inflammation, intestinal barrier and gut microbiota. CC may be an effective and safe therapeutic choice for STC.
Collapse
Affiliation(s)
- Xiaoyu Gao
- Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming, China
| | - Yifan Hu
- Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming, China
- College of Agronomy and Biotechnology, Yunnan Agricultural University, Kunming, China
| | - Yafei Tao
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Shuangfeng Liu
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Haowen Chen
- College of Agronomy and Biotechnology, Yunnan Agricultural University, Kunming, China
| | - Jiayi Li
- College of Agronomy and Biotechnology, Yunnan Agricultural University, Kunming, China
| | - Yan Zhao
- Department of Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Jun Sheng
- Yunnan Provincial Engineering Research Center for Edible and Medicinal Homologous Functional Food, Yunnan Agricultural University, Kunming, China
- *Correspondence: Jun Sheng,
| | - Yang Tian
- Yunnan Provincial Engineering Research Center for Edible and Medicinal Homologous Functional Food, Yunnan Agricultural University, Kunming, China
- Yang Tian,
| | - Yuanhong Fan
- Yunnan Plateau Characteristic Agricultural Industry Research Institute, Yunnan Agricultural University, Kunming, China
- Yuanhong Fan,
| |
Collapse
|
15
|
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: 2.3] [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
|
16
|
Zhang Q, Zhou S, Lim PE, Wei B, Xue C, Xue Y, Tang Q. Kappaphycus Alvarezii Compound Powder Prevents Chemotherapy-Induced Intestinal Mucositis in BALB/c Mice. Nutr Cancer 2022; 74:3735-3746. [PMID: 35758096 DOI: 10.1080/01635581.2022.2089699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
This study aimed to formulate Kappaphycus alvarezii compound powder containing Kappaphycus alvarezii powder (KP), cooked sorghum powder (SP), and longan powder (LP); which was evaluated for its therapeutic effects against chemotherapy-induced intestinal mucosal injury (CIMI). Based on rheological properties, sensory evaluation, and antioxidant activity and using single factor and response surface methodology, the optimal formula to develop the compound powder was determined to be 35% KP, 30% SP, 5% LP, and 30% xylitol. Thereafter, the efficacy of the compound powder was tested by feeding BALB/c mice with diets supplemented with the Kappaphycus alvarezii compound powder (3% and 5%) for 14 consecutive days. The chemotherapeutic drug 5-fluorouracil was intraperitoneally injected (50 mg/kg) in the mice to induce CIMI for the last three consecutive days. Compared to the CIMI mice, those fed 5% Kappaphycus alvarezii compound powder (HC) showed significantly improved the intestinal injury, increased mucin-2 secretion, and reduced TNF-α, IL-1β, IL-6, LT, and COX-2 levels. Furthermore, HC intake significantly reduced the Firmicutes-to-Bacteroidetes ratio, promoted the growth of beneficial bacteria, such as Alloprevotella, and inhibited the growth of harmful bacteria, such as Clostridium. In conclusion, HC has a protective effect against CIMI and provides a novel dietary strategy for patients undergoing chemotherapy.
Collapse
Affiliation(s)
- Qing Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Sainan Zhou
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Phaik Eem Lim
- Institute of Ocean and Earth Sciences, University of Malaya, Kuala Lumpur, Malaysia
| | - Biqian Wei
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Changhu Xue
- College of Food Science and Engineering, Ocean University of China, Qingdao, China.,Pilot National Laboratory for Marine Science and Technology, Laboratory for Marine Drugs and Bioproducts, Qingdao, China
| | - Yong Xue
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Qingjuan Tang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| |
Collapse
|
17
|
Guilloteau E, Djouina M, Caboche S, Waxin C, Deboudt K, Beury D, Hot D, Pichavant M, Dubuquoy L, Launay D, Vignal C, Choël M, Body-Malapel M. Exposure to atmospheric Ag, TiO 2, Ti and SiO 2 engineered nanoparticles modulates gut inflammatory response and microbiota in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 236:113442. [PMID: 35367877 DOI: 10.1016/j.ecoenv.2022.113442] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 03/08/2022] [Accepted: 03/19/2022] [Indexed: 06/14/2023]
Abstract
The development of nanotechnologies is leading to greater abundance of engineered nanoparticles (EN) in the environment, including in the atmospheric air. To date, it has been shown that the most prevalent EN found in the air are silver (Ag), titanium dioxide (TiO2), titanium (Ti), and silicon dioxide (SiO2). As the intestinal tract is increasingly recognized as a target for adverse effects induced by inhalation of air particles, the aim of this study was to assess the impact of these 4 atmospheric EN on intestinal inflammation and microbiota. We assessed the combined toxicity effects of Ag, Ti, TiO2, and SiO2 following a 28-day inhalation protocol in male and female mice. In distal and proximal colon, and in jejunum, EN mixture inhalation did not induce overt histological damage, but led to a significant modulation of inflammatory cytokine transcript abundance, including downregulation of Tnfα, Ifnγ, Il1β, Il17a, Il22, IL10, and Cxcl1 mRNA levels in male jejunum. A dysbiosis was observed in cecal microbiota of male and female mice exposed to the EN mixture, characterized by sex-dependent modulations of specific bacterial taxa, as well as sex-independent decreased abundance of the Eggerthellaceae family. Under dextran sodium sulfate-induced inflammatory conditions, exposure to the EN mixture increased the development of colitis in both male and female mice. Moreover, the direct dose-response effects of individual and mixed EN on gut organoids was studied and Ag, TiO2, Ti, SiO2, and EN mixture were found to generate specific inflammatory responses in the intestinal epithelium. These results indicate that the 4 most prevalent atmospheric EN could have the ability to disturb intestinal homeostasis through direct modulation of cytokine expression in gut epithelium, and by altering the inflammatory response and microbiota composition following inhalation.
Collapse
Affiliation(s)
- Eva Guilloteau
- University of Lille, Inserm, CHU Lille, U1286- INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Madjid Djouina
- University of Lille, Inserm, CHU Lille, U1286- INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Ségolène Caboche
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR2014 - US41 - PLBS-Plateformes Lilloises de Biologie & Santé, F-59000 Lille, France
| | - Christophe Waxin
- University of Lille, Inserm, CHU Lille, U1286- INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Karine Deboudt
- Université du Littoral Côte d'Opale, EA 4493 - LPCA - Laboratoire de Physico-Chimie de l'Atmosphère, 59140 Dunkerque, France
| | - Delphine Beury
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR2014 - US41 - PLBS-Plateformes Lilloises de Biologie & Santé, F-59000 Lille, France
| | - David Hot
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR2014 - US41 - PLBS-Plateformes Lilloises de Biologie & Santé, F-59000 Lille, France
| | - Muriel Pichavant
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Laurent Dubuquoy
- University of Lille, Inserm, CHU Lille, U1286- INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - David Launay
- University of Lille, Inserm, CHU Lille, U1286- INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Cécile Vignal
- University of Lille, Inserm, CHU Lille, U1286- INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Marie Choël
- University of Lille, CNRS, UMR 8516 - LASIRE - Laboratoire de Spectroscopie pour les Interactions, la Réactivité et l'Environnement, F-59000 Lille, France
| | - Mathilde Body-Malapel
- University of Lille, Inserm, CHU Lille, U1286- INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France.
| |
Collapse
|
18
|
Sun Y, Ding X, Cui Y, Li H, Wang D, Liang H, Liu S, Zhang X, Wang H, Sun T. Positive Effects of Neutrophil Elastase Inhibitor (Sivelestat) on Gut Microbiome and Metabolite Profiles of Septic Rats. Front Cell Infect Microbiol 2022; 12:818391. [PMID: 35372122 PMCID: PMC8965314 DOI: 10.3389/fcimb.2022.818391] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 02/17/2022] [Indexed: 12/22/2022] Open
Abstract
Background Neutrophil elastase (NE) is associated with sepsis occurrence and progression. We hypothesized that the NE inhibitor Sivelestat might modulate abnormal gut microbiota and metabolites during sepsis. Methods Sixty Sprague-Dawley (SD) rats were randomly divided into sham control (SC), sepsis (CLP), and sepsis+Sivelestat (Sive) groups. The rats’ survival status was monitored for 24 hours postoperatively, and feces were collected for microbiome and non-targeted metabolomics analyses. Results Sivelestat administration significantly improved the survival of septic rats (80% vs 50%, P = 0.047). Microbiome analysis showed that the microbiota composition of rats in the CLP group was significantly disturbed, as potential pathogens such as Escherichia-Shigella and Gammaproteobacteria became dominant, and the beneficial microbiota represented by Lactobacillus decreased. These changes were reversed in Sive group, and the overall microbial status was restored to a similar composition to SC group. Differential analysis identified 36 differential operational taxonomic units and 11 metabolites between the Sive and CLP groups, such as 6-Aminopenicillanic acid, gamma-Glutamyl-leucine, and cortisone (variable importance in projection>1and P<0.05). These discriminatory metabolites were highly correlated with each other and mainly involved in the phenylalanine, tyrosine, and tryptophan biosynthesis pathways. Integrated microbiome and metabolome analyses found that almost all Sivelestat-modulated microbes were associated with differential metabolites (P < 0.05), such as Lactobacillus and some amino acids, suggesting that the Sivelestat-induced metabolic profile differences were in part due to its influence on the gut microbiome. Conclusion Sivelestat administration in septic rats improved survival, gut microbiota composition and associated metabolites, which could provide new options for sepsis treatment.
Collapse
|
19
|
Cloud AS, Vargheese AM, Gunewardena S, Shimak RM, Ganeshkumar S, Kumaraswamy E, Jensen RA, Chennathukuzhi VM. Loss of REST in breast cancer promotes tumor progression through estrogen sensitization, MMP24 and CEMIP overexpression. BMC Cancer 2022; 22:180. [PMID: 35177031 PMCID: PMC8851790 DOI: 10.1186/s12885-022-09280-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 02/08/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Breast cancer is the most common malignancy in women, and is both pathologically and genetically heterogeneous, making early detection and treatment difficult. A subset of breast cancers express normal levels of REST (repressor element 1 silencing transcription factor) mRNA but lack functional REST protein. Loss of REST function is seen in ~ 20% of breast cancers and is associated with a more aggressive phenotype and poor prognosis. Despite the frequent loss of REST, little is known about the role of REST in the molecular pathogenesis of breast cancer. METHODS TCGA data was analyzed for the expression of REST target genes in breast cancer patient samples. We then utilized gene knockdown in MCF-7 cells in the presence or absence of steroid hormones estrogen and/ progesterone followed by RNA sequencing, as well as chromatin immunoprecipitation and PCR in an attempt to understand the tumor suppressor role of REST in breast cancer. RESULTS We show that REST directly regulates CEMIP (cell migration-inducing and hyaluronan-binding protein, KIAA1199) and MMP24 (matrix metallopeptidase 24), genes known to have roles in invasion and metastasis. REST knockdown in breast cancer cells leads to significant upregulation of CEMIP and MMP24. In addition, we found REST binds to RE-1 sites (repressor element-1) within the genes and influences their transcription. Furthermore, we found that the estrogen receptor (ESR1) signaling pathway is activated in the absence of REST, regardless of hormone treatment. CONCLUSIONS We demonstrate a critical role for the loss of REST in aggressive breast cancer pathogenesis and provide evidence for REST as an important diagnostic marker for personalized treatment plans.
Collapse
Affiliation(s)
- Ashley S. Cloud
- grid.412016.00000 0001 2177 6375Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS USA
| | - Aditya M. Vargheese
- grid.412016.00000 0001 2177 6375Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS USA ,grid.468219.00000 0004 0408 2680The University of Kansas Cancer Center, Kansas City, KS USA ,grid.266515.30000 0001 2106 0692University of Kansas, Lawrence, KS USA
| | - Sumedha Gunewardena
- grid.412016.00000 0001 2177 6375Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS USA ,grid.412016.00000 0001 2177 6375Department of Biostatistics, University of Kansas Medical Center, Kansas City, KS USA
| | - Raeann M. Shimak
- grid.468219.00000 0004 0408 2680The University of Kansas Cancer Center, Kansas City, KS USA ,grid.412016.00000 0001 2177 6375Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS USA
| | - Sornakala Ganeshkumar
- grid.412016.00000 0001 2177 6375Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS USA
| | - Easwari Kumaraswamy
- grid.468219.00000 0004 0408 2680The University of Kansas Cancer Center, Kansas City, KS USA ,grid.412016.00000 0001 2177 6375Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS USA
| | - Roy A. Jensen
- grid.468219.00000 0004 0408 2680The University of Kansas Cancer Center, Kansas City, KS USA ,grid.266515.30000 0001 2106 0692University of Kansas, Lawrence, KS USA ,grid.412016.00000 0001 2177 6375Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS USA ,grid.412016.00000 0001 2177 6375Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS USA ,grid.412016.00000 0001 2177 6375Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS USA
| | - Vargheese M. Chennathukuzhi
- grid.412016.00000 0001 2177 6375Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS USA ,grid.468219.00000 0004 0408 2680The University of Kansas Cancer Center, Kansas City, KS USA
| |
Collapse
|
20
|
Sheng K, Xu Y, Kong X, Wang J, Zha X, Wang Y. Probiotic Bacillus cereus Alleviates Dextran Sulfate Sodium-Induced Colitis in Mice through Improvement of the Intestinal Barrier Function, Anti-Inflammation, and Gut Microbiota Modulation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:14810-14823. [PMID: 34677958 DOI: 10.1021/acs.jafc.1c03375] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Dysbiosis leads to continuous progress of inflammatory bowel disease (IBD). However, current therapeutic approaches for IBD have limited efficacy and are associated with various side effects. This study focused on exploring the positive effect of a new Bacillus cereus (B. cereus) strain (HMPM18123) in a colitis mouse model and elucidate the underlying molecular mechanisms. The colitis symptoms were alleviated by the B. cereus administration as evidenced by decreased body weight loss, colon length shortening, disease activity index score, and histopathological score. The B. cereus mitigated intestinal epithelial barrier damage by upregulating tight junction protein expression. Moreover, B. cereus exerted anti-inflammatory effects by regulating macrophage polarization and suppressing the TLR4-NF-κB-NLRP3 inflammasome signaling pathways. B. cereus also rebalanced the damaged gut microbiota. Thus, the molecular mechanism of alleviating colitis by B. cereus treatment involved the regulation of the TLR4-NF-κB-NLRP3 inflammasome signaling pathways in intestinal mucosal barriers by modulating gut microbiota composition.
Collapse
Affiliation(s)
- Kangliang Sheng
- School of Life Sciences, Anhui University, Hefei, Anhui 230601, China
- Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, Anhui 230601, China
- Anhui Key Laboratory of Modern Biomanufacturing, Anhui University, Hefei, Anhui 230601, China
| | - Yifan Xu
- School of Life Sciences, Anhui University, Hefei, Anhui 230601, China
- Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, Anhui 230601, China
- Anhui Key Laboratory of Modern Biomanufacturing, Anhui University, Hefei, Anhui 230601, China
| | - Xiaowei Kong
- School of Life Sciences, Anhui University, Hefei, Anhui 230601, China
- Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, Anhui 230601, China
- Anhui Key Laboratory of Modern Biomanufacturing, Anhui University, Hefei, Anhui 230601, China
| | - Jingmin Wang
- School of Life Sciences, Anhui University, Hefei, Anhui 230601, China
- Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, Anhui 230601, China
- Anhui Key Laboratory of Modern Biomanufacturing, Anhui University, Hefei, Anhui 230601, China
| | - Xiangdong Zha
- School of Life Sciences, Anhui University, Hefei, Anhui 230601, China
- Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, Anhui 230601, China
- Anhui Key Laboratory of Modern Biomanufacturing, Anhui University, Hefei, Anhui 230601, China
| | - Yongzhong Wang
- School of Life Sciences, Anhui University, Hefei, Anhui 230601, China
- Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, Anhui 230601, China
- Anhui Key Laboratory of Modern Biomanufacturing, Anhui University, Hefei, Anhui 230601, China
- Institute of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China
| |
Collapse
|
21
|
Zhang T, Zhang S, Jin C, Lin Z, Deng T, Xie X, Deng L, Li X, Ma J, Ding X, Liu Y, Shan Y, Yu Z, Wang Y, Chen G, Li J. A Predictive Model Based on the Gut Microbiota Improves the Diagnostic Effect in Patients With Cholangiocarcinoma. Front Cell Infect Microbiol 2021; 11:751795. [PMID: 34888258 PMCID: PMC8650695 DOI: 10.3389/fcimb.2021.751795] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 11/01/2021] [Indexed: 12/12/2022] Open
Abstract
Cholangiocarcinoma (CCA) is a malignant hepatic tumor with a poor prognosis, which needs early diagnosis urgently. The gut microbiota has been shown to play a crucial role in the progression of liver cancer. Here, we explored a gut microbiota model covering genera Burkholderia-Caballeronia-Paraburkholderia, Faecalibacterium, and Ruminococcus_1 (B-F-R) for CCA early diagnosis. A case-control study was conducted to enroll 53 CCA patients, 47 cholelithiasis patients, and 40 healthy controls. The feces samples and clinical information of participants were collected in the same period. The gut microbiota and its diversity of individuals were accessed with 16S rDNA sequencing, and the gut microbiota profile was evaluated according to microbiota diversity. Finally, four enriched genera in the CCA group (genera Bacteroides, Muribaculaceae_unclassified, Muribaculum, and Alistipes) and eight enriched genera in the cholelithiasis group (genera Bifidobacterium, Streptococcus, Agathobacter, Ruminococcus_gnavus_group, Faecalibacterium, Subdoligranulum, Collinsella, Escherichia-Shigella) constitute an overall different microbial community composition (P = 0.001). The B-F-R genera model with better diagnostic value than carbohydrate antigen 19-9 (CA19-9) was identified by random forest and Statistical Analysis of Metagenomic Profiles (STAMP) to distinguish CCA patients from healthy controls [area under the curve (AUC) = 0.973, 95% CI = 0.932–1.0]. Moreover, the correlative analysis found that genera Burkholderia-Caballeronia-Paraburkholderia were positively correlated with body mass index (BMI). The significantly different microbiomes between cholelithiasis and CCA were found via principal coordinates analysis (PCoA) and linear discriminant analysis effect size (LEfSe), and Venn diagram and LEfSe were utilized to identify four genera by comparing microbial compositions among patients with malignant obstructive jaundice (MOJ-Y) or not (MOJ-N). In brief, our findings suggest that gut microbiota vary from benign and malignant hepatobiliary diseases to healthy people and provide evidence supporting gut microbiota to be a non-invasive biomarker for the early diagnosis of CCA.
Collapse
Affiliation(s)
- Tan Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Sina Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chen Jin
- Department of Epidemiology and Biostatistics, School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Zixia Lin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Tuo Deng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaozai Xie
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Liming Deng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xueyan Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jun Ma
- Department of Epidemiology and Biostatistics, School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Xiwei Ding
- Department of Gastroenterology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yaming Liu
- Department of Gastroenterology, Zhongshan Hospital Xiamen University, Xiamen, China
| | - Yunfeng Shan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhengping Yu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yi Wang
- Department of Epidemiology and Biostatistics, School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Gang Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jialiang Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
22
|
Jakimiuk K, Gesek J, Atanasov AG, Tomczyk M. Flavonoids as inhibitors of human neutrophil elastase. J Enzyme Inhib Med Chem 2021; 36:1016-1028. [PMID: 33980119 PMCID: PMC8128182 DOI: 10.1080/14756366.2021.1927006] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/28/2021] [Accepted: 05/02/2021] [Indexed: 12/17/2022] Open
Abstract
Elastase is a proteolytic enzyme belonging to the family of hydrolases produced by human neutrophils, monocytes, macrophages, and endothelial cells. Human neutrophil elastase is known to play multiple roles in the human body, but an increase in its activity may cause a variety of diseases. Elastase inhibitors may prevent the development of psoriasis, chronic kidney disease, respiratory disorders (including COVID-19), immune disorders, and even cancers. Among polyphenolic compounds, some flavonoids and their derivatives, which are mostly found in herbal plants, have been revealed to influence elastase release and its action on human cells. This review focuses on elastase inhibitors that have been discovered from natural sources and are biochemically characterised as flavonoids. The inhibitory activity on elastase is a characteristic of flavonoid aglycones and their glycoside and methylated, acetylated and hydroxylated derivatives. The presented analysis of structure-activity relationship (SAR) enables the determination of the chemical groups responsible for evoking an inhibitory effect on elastase. Further study especially of the in vivo efficacy and safety of the described natural compounds is of interest in order to gain better understanding of their health-promoting potential.
Collapse
Affiliation(s)
- Katarzyna Jakimiuk
- Department of Pharmacognosy, Faculty of Pharmacy with the Division of Laboratory Medicine, Medical University of Białystok, Białystok, Poland
| | - Jakub Gesek
- Department of Pharmacognosy, Medical University of Białystok, Student’s Scientific Association, Białystok, Poland
| | - Atanas G. Atanasov
- Ludwig Boltzmann Institute for Digital Health and Patient Safety, Medical University of Vienna, Vienna, Austria
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzębiec, Poland
- Department of Pharmacognosy, University of Vienna, Vienna, Austria
| | - Michał Tomczyk
- Department of Pharmacognosy, Faculty of Pharmacy with the Division of Laboratory Medicine, Medical University of Białystok, Białystok, Poland
| |
Collapse
|
23
|
Human Antibody Domains and Fragments Targeting Neutrophil Elastase as Candidate Therapeutics for Cancer and Inflammation-Related Diseases. Int J Mol Sci 2021; 22:ijms222011136. [PMID: 34681796 PMCID: PMC8539514 DOI: 10.3390/ijms222011136] [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: 09/29/2021] [Revised: 10/10/2021] [Accepted: 10/12/2021] [Indexed: 11/25/2022] Open
Abstract
Neutrophil elastase (NE) is a serine protease released during neutrophil maturation. High levels of NE are related to lung tissue damage and poor prognosis in cancer; thus, NE is a potential target for therapeutic immunotherapy for multiple lung diseases and cancers. Here, we isolate and characterize two high-affinity, specific, and noncompetitive anti-NE antibodies Fab 1C10 and VH 1D1.43 from two large phage-displayed human Fab and VH libraries. After fusion with human IgG1 Fc, both of them (VH-Fc 1D1.43 and IgG1 1C10) inhibit NE enzymatic activity with VH-Fc 1D1.43 showing comparable inhibitory effects to that of the small molecule NE inhibitor SPCK and IgG1 1C10 exhibiting even higher (2.6-fold) activity than SPCK. Their epitopes, as mapped by peptide arrays combined with structural modeling, indicate different mechanisms for blocking NE activity. Both VH-Fc and IgG1 antibodies block NE uptake by cancer cells and fibroblast differentiation. VH-Fc 1D1.43 and IgG1 1C10 are promising for the antibody-based immunotherapy of cancer and inflammatory diseases.
Collapse
|
24
|
Mun Y, Hwang JS, Shin YJ. Role of Neutrophils on the Ocular Surface. Int J Mol Sci 2021; 22:10386. [PMID: 34638724 PMCID: PMC8508808 DOI: 10.3390/ijms221910386] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/16/2021] [Accepted: 09/23/2021] [Indexed: 02/07/2023] Open
Abstract
The ocular surface is a gateway that contacts the outside and receives stimulation from the outside. The corneal innate immune system is composed of many types of cells, including epithelial cells, fibroblasts, natural killer cells, macrophages, neutrophils, dendritic cells, mast cells, basophils, eosinophils, mucin, and lysozyme. Neutrophil infiltration and degranulation occur on the ocular surface. Degranulation, neutrophil extracellular traps formation, called NETosis, and autophagy in neutrophils are involved in the pathogenesis of ocular surface diseases. It is necessary to understand the role of neutrophils on the ocular surface. Furthermore, there is a need for research on therapeutic agents targeting neutrophils and neutrophil extracellular trap formation for ocular surface diseases.
Collapse
Affiliation(s)
- Yongseok Mun
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07442, Korea; (Y.M.); (J.S.H.)
- Hallym BioEyeTech Research Center, Hallym University College of Medicine, Seoul 07442, Korea
| | - Jin Sun Hwang
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07442, Korea; (Y.M.); (J.S.H.)
- Hallym BioEyeTech Research Center, Hallym University College of Medicine, Seoul 07442, Korea
| | - Young Joo Shin
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07442, Korea; (Y.M.); (J.S.H.)
- Hallym BioEyeTech Research Center, Hallym University College of Medicine, Seoul 07442, Korea
| |
Collapse
|
25
|
Zhang Y, Liu W, Zhang D, Yang Y, Wang X, Li L. Fermented and Germinated Processing Improved the Protective Effects of Foxtail Millet Whole Grain Against Dextran Sulfate Sodium-Induced Acute Ulcerative Colitis and Gut Microbiota Dysbiosis in C57BL/6 Mice. Front Nutr 2021; 8:694936. [PMID: 34395495 PMCID: PMC8358663 DOI: 10.3389/fnut.2021.694936] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/07/2021] [Indexed: 12/30/2022] Open
Abstract
This study investigated the effects of foxtail millet whole grain flours obtained through different processing methods on alleviating symptoms and gut microbiota dysbiosis in a dextran sulfate sodium (DSS)-induced murine colitis model. Sixty C57BL/6 mice were divided into six groups (n = 10 in each group), including one control group (CTRL) without DSS treatment and five DSS-treated groups receiving one of the following diets: AIN-93M standard diet (93MD), whole grain foxtail millet flour (FM), fermented (F-FM), germinated (G-FM), and fermented-germinated foxtail millet flour (FG-FM). A comparison of the disease activity index (DAI) demonstrated that foxtail millet whole grain-based diets could alleviate the symptoms of enteritis to varying degrees. In addition, 16S rRNA gene sequencing revealed that FG-FM almost completely alleviated DSS-induced dysbiosis. Mice on the FG-FM diet also had the lowest plasma IL-6 levels and claudin2 expression levels in the colon, indicating reduced systemic inflammation and improved gut barrier function. This study suggested that foxtail millet whole grain is an attractive choice for the intervention of IBD and gut microbiota dysbiosis, and its prebiotic properties are highly affected by the processing methods.
Collapse
Affiliation(s)
- Yuhan Zhang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China.,Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan, China.,Qilu Hospital, Shandong University, Jinan, China
| | - Wei Liu
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Di Zhang
- Qilu Hospital, Shandong University, Jinan, China
| | - Yanbing Yang
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Xianshu Wang
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Lingfei Li
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
| |
Collapse
|