1
|
Kim SH, Lee R, Yoon JW, Cheong HT, Ra CS, Rhee KJ, Park J, Jung BD. Anti-Inflammatory Effects of Zinc Oxide and Berberine in Rats with Dextran Sulfate Sodium (DSS)-Induced Colitis. Animals (Basel) 2024; 14:1919. [PMID: 38998031 PMCID: PMC11240726 DOI: 10.3390/ani14131919] [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: 05/14/2024] [Revised: 06/14/2024] [Accepted: 06/27/2024] [Indexed: 07/14/2024] Open
Abstract
Zinc oxide (ZnO) is frequently used in high concentrations to prevent diarrhea in weaning pigs. However, it can produce environmental pollution, because it is not absorbed by the intestines and is excreted in the feces. In studies to identify an alternative substance to ZnO, we used a model of colitis induced by dextran sulfate sodium (DSS) in rats to compare the anti-inflammatory effects of berberine with ZnO. DSS-treated rats displayed weight loss, shortening of the colon, increased fecal water content, and an increase in the disease activity index (DAI). In contrast, DSS + ZnO- and DSS + berberine-treated rats exhibited reduced colon shortening, decreased fecal water content, and a decrease in the DAI. Histological analysis revealed that both ZnO and berberine treatment reduced epithelial cell damage, crypt destruction, and infiltration of inflammatory cells. Moreover, the liver damage index was not significantly different between ZnO and berberine-treated rats. This study indicated that both ZnO and berberine can improve DSS-induced colitis in rats and suggests berberine as an alternative treatment to ZnO that would not cause environmental pollution.
Collapse
Affiliation(s)
- Seon-Hyoung Kim
- College of Veterinary Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Rangyeon Lee
- College of Veterinary Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
- Multidimensional Genomics Research Center, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Jang-Won Yoon
- College of Veterinary Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
- Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Hee-Tae Cheong
- College of Veterinary Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
- Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Chang-Six Ra
- College of Animal Life Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Ki-Jong Rhee
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University Mirae Campus, Wonju 26493, Republic of Korea
| | - Jeongho Park
- College of Veterinary Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
- Multidimensional Genomics Research Center, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Bae-Dong Jung
- College of Veterinary Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
- Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| |
Collapse
|
2
|
Zhang Y, Wu Z, Zhao Q, Liu Y, Huang Q, Zhang M, Li S, Wang D, Li N, Chi Y, Liu Y. Mesenteric Lymphatic B Cells Migrate to the Intestine and Aggravate DSS-Induced Colitis via the CXCR5-CXCL13 Axis. BIOLOGY 2024; 13:322. [PMID: 38785804 PMCID: PMC11117591 DOI: 10.3390/biology13050322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/29/2024] [Accepted: 05/02/2024] [Indexed: 05/25/2024]
Abstract
The pathogenesis of inflammatory bowel disease (IBD) is still unknown. Mesenteric lymphatics (MLs), which are closely related to the intestine in both anatomy and physiology, have been suggested to be involved in IBD. In the present study, we aim to investigate the effects of ML immune cells on IBD and explore the potential associated mechanisms. Acute colitis was induced in rats using dextran sulfate sodium salt (DSS). Mesenteric lymphangiogenesis, ML stenosis, and dilation were observed, with an increased proportion of MLB cells in DSS-induced colitis rats. The adoptive transfer of B cells isolated from ML (MLB) was employed to investigate their effects on colitis. MLB cells derived from DSS-induced colitis rats exhibited a higher propensity to migrate to the intestine. The proportion of colonic T cells was altered, along with the aggravated colitis induced by the adoptive transfer of MLB cells derived from DSS-induced colitis rats. RNA sequencing revealed increased Cxcr5 expression in MLB cells from colitis rats, while real-time PCR indicated an upregulation of its ligand Cxcl13 in the colon of colitis rats. These findings suggest that MLB cells may migrate to the intestine and aggravate colitis. In summary, colonic T cells respond to MLB cells from colitis rats, and MLB cells aggravate DSS-induced colitis via the CXCR5-CXCL13 axis.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Gastroenterology, Peking University People’s Hospital, Beijing 100044, China; (Y.Z.); (Z.W.); (Q.Z.)
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People’s Hospital, Beijing 100044, China
| | - Zhe Wu
- Department of Gastroenterology, Peking University People’s Hospital, Beijing 100044, China; (Y.Z.); (Z.W.); (Q.Z.)
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People’s Hospital, Beijing 100044, China
| | - Qinghe Zhao
- Department of Gastroenterology, Peking University People’s Hospital, Beijing 100044, China; (Y.Z.); (Z.W.); (Q.Z.)
- Department of Central Laboratory and Institute of Clinical Molecular Biology, Peking University People’s Hospital, Beijing 100044, China; (D.W.); (N.L.)
| | - Yaming Liu
- Department of Gastroenterology and Hepatology, Xiamen University Zhongshan Hospital, Xiamen 361001, China;
| | - Qing Huang
- Department of Gastroenterology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China;
| | - Menglei Zhang
- Department of Animal Laboratory, Peking University People’s Hospital, Beijing 100044, China; (M.Z.); (S.L.)
| | - Shuolei Li
- Department of Animal Laboratory, Peking University People’s Hospital, Beijing 100044, China; (M.Z.); (S.L.)
| | - Di Wang
- Department of Central Laboratory and Institute of Clinical Molecular Biology, Peking University People’s Hospital, Beijing 100044, China; (D.W.); (N.L.)
| | - Na Li
- Department of Central Laboratory and Institute of Clinical Molecular Biology, Peking University People’s Hospital, Beijing 100044, China; (D.W.); (N.L.)
| | - Yujing Chi
- Department of Gastroenterology, Peking University People’s Hospital, Beijing 100044, China; (Y.Z.); (Z.W.); (Q.Z.)
- Department of Central Laboratory and Institute of Clinical Molecular Biology, Peking University People’s Hospital, Beijing 100044, China; (D.W.); (N.L.)
| | - Yulan Liu
- Department of Gastroenterology, Peking University People’s Hospital, Beijing 100044, China; (Y.Z.); (Z.W.); (Q.Z.)
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People’s Hospital, Beijing 100044, China
| |
Collapse
|
3
|
Zhou YW, Ren Y, Lu MM, Xu LL, Cheng WX, Zhang MM, Ding LP, Chen D, Gao JG, Du J, Jin CL, Chen CX, Li YF, Cheng T, Jiang PL, Yang YD, Qian PX, Xu PF, Jin X. Crohn's disease as the intestinal manifestation of pan-lymphatic dysfunction: An exploratory proposal based on basic and clinical data. World J Gastroenterol 2024; 30:34-49. [PMID: 38293325 PMCID: PMC10823898 DOI: 10.3748/wjg.v30.i1.34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/08/2023] [Accepted: 12/27/2023] [Indexed: 01/06/2024] Open
Abstract
Crohn's disease (CD) is caused by immune, environmental, and genetic factors. It can involve the entire gastrointestinal tract, and although its prevalence is rapidly increasing its etiology remains unclear. Emerging biological and small-molecule drugs have advanced the treatment of CD; however, a considerable proportion of patients are non-responsive to all known drugs. To achieve a breakthrough in this field, innovations that could guide the further development of effective therapies are of utmost urgency. In this review, we first propose the innovative concept of pan-lymphatic dysfunction for the general distribution of lymphatic dysfunction in various diseases, and suggest that CD is the intestinal manifestation of pan-lymphatic dysfunction based on basic and clinical preliminary data. The supporting evidence is fully summarized, including the existence of lymphatic system dysfunction, recognition of the inside-out model, disorders of immune cells, changes in cell plasticity, partial overlap of the underlying mechanisms, and common gut-derived fatty and bile acid metabolism. Another benefit of this novel concept is that it proposes adopting the zebrafish model for studying intestinal diseases, especially CD, as this model is good at presenting and mimicking lymphatic dysfunction. More importantly, the ensuing focus on improving lymphatic function may lead to novel and promising therapeutic strategies for CD.
Collapse
Affiliation(s)
- Yu-Wei Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Yue Ren
- Department of Gastroenterology, The Second Hospital of Jiaxing, Jiaxing 314000, Zhejiang Province, China
| | - Miao-Miao Lu
- Endoscopy Center, Children’s Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Ling-Ling Xu
- Department of Gastroenterology, The Second People’s Hospital of Yuhang District, Hangzhou 310000, Zhejiang Province, China
| | - Wei-Xin Cheng
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Meng-Meng Zhang
- Department of Gastroenterology, Hangzhou Shangcheng District People’s Hospital, Hangzhou 310003, Zhejiang Province, China
| | - Lin-Ping Ding
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Dong Chen
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Jian-Guo Gao
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Juan Du
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Ci-Liang Jin
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| | - Chun-Xiao Chen
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Yun-Fei Li
- Women’s Hospital and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Tao Cheng
- Women’s Hospital and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Peng-Lei Jiang
- Center of Stem Cell and Regenerative Medicine, and Bone Marrow Transplantation Center, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Yi-Da Yang
- Department of Infectious Disease, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Peng-Xu Qian
- Center of Stem Cell and Regenerative Medicine, and Bone Marrow Transplantation Center, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Peng-Fei Xu
- Women’s Hospital and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Xi Jin
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| |
Collapse
|
4
|
Carrillo Diaz de Leon M, Keane K, Roizes S, Liao S, von der Weid PY, Stephens M. Not just fibrotic: endothelial-derived TGFβ maintains contractile function and lymphatic muscle phenotype during homeostasis. Am J Physiol Cell Physiol 2024; 326:C269-C281. [PMID: 38047303 DOI: 10.1152/ajpcell.00327.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 11/07/2023] [Accepted: 11/22/2023] [Indexed: 12/05/2023]
Abstract
Cell-cell communication within the lymphatic vasculature during homeostasis is incompletely detailed. Although many discoveries highlight the pathological roles of transforming growth factor-beta (TGFβ) in chronic vascular inflammation and associated fibrosis, only a small amount is known surrounding the role of TGFβ-signaling in homeostatic lymphatic function. Here, we discovered that pharmacological blockade of TGFβ receptor 1 (TGFβR1) negatively impacts rat mesenteric lymphatic vessel pumping, significantly reducing vessel contractility and surrounding lymphatic muscle coverage. We have identified mesenteric lymphatic endothelial cells themselves as a source of endogenous vascular TGFβ and that TGFβ production is significantly increased in these cells via activation of a number of functional pattern recognition receptors they express. We show that a continuous supply of TGFβ is essential to maintain the contractile phenotype of neighboring lymphatic muscle cells and support this conclusion through in vitro analysis of primary isolated lymphatic muscle cells that undergo synthetic differentiation during 2-D cell culture, a phenomenon that could be effectively rescued by supplementation with recombinant TGFβ. Finally, we demonstrate that lymphatic endothelial production of TGFβ is regulated, in part, by nitric oxide in a manner we propose is essential to counteract the pathological over-production of TGFβ. Taken together, these data highlight the essential role of homeostatic TGFβ signaling in the maintenance of lymphatic vascular function and highlight possible deleterious consequences of its inhibition.NEW & NOTEWORTHY The growth factor TGFβ is commonly associated with its pathological overproduction during tissue fibrosis rather than its homeostatic functions. We expose the lymphatic endothelium as a source of endogenous TGFβ, the impact of its production on the maintenance of surrounding lymphatic muscle cell phenotype, and internally regulated mechanisms of its production. Overall, these results highlight the intricate balance of TGFβ-signaling as an essential component of maintaining lymphatic contractile function.
Collapse
Affiliation(s)
- Miriam Carrillo Diaz de Leon
- Inflammation Research Network, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Department of Microbiology, Immunology & Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Keith Keane
- Inflammation Research Network, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Simon Roizes
- Inflammation Research Network, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Shan Liao
- Inflammation Research Network, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Department of Microbiology, Immunology & Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Pierre-Yves von der Weid
- Inflammation Research Network, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Matthew Stephens
- Inflammation Research Network, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| |
Collapse
|
5
|
Juneja P, Sharma A, Shasthry SM, Kumar G, Tripathi DM, Rajan V, Rastogi A, Sarin SK, Kaur S. Podoplanin-positive dilated lymphatic vessels in duodenum associates with three-month mortality in patients with cirrhosis. Front Physiol 2023; 14:1045983. [PMID: 37304826 PMCID: PMC10248415 DOI: 10.3389/fphys.2023.1045983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 05/16/2023] [Indexed: 06/13/2023] Open
Abstract
Dilated and dysfunctional gut lymphatic vessels (LVs) have been reported in experimental cirrhosis. Here, we studied LVs in duodenal (D2)-biopsies of liver cirrhosis patients and investigated the prognostic role of a LV marker, podoplanin (PDPN), in predicting the mortality of patients with cirrhosis. A prospective, single-center cohort study was performed in liver cirrhosis patients (n = 31) and matched healthy controls (n = 9). D2-biopsies were obtained during endoscopy procedure, immunostained with PDPN, and scored based on 1) intensity and 2) density of positively-stained LVs per high power field. Gut and systemic inflammation were estimated by quantifying duodenal CD3+ intraepithelial lymphocytes (IELs), CD68+ macrophages, and serum TNF-α and IL-6 levels, respectively. Gut permeability and inflammation as assessed by quantifying gene expression of TJP1, OCLN, TNF-α, and IL-6 in D2-biopsies. Gene expression of LV markers, PDPN (8-fold), and LYVE1 (3-fold) was enhanced in D2-biopsies of cirrhosis patients compared to control (p < 0.0001). The mean PDPN score in decompensated cirrhosis patients (6.91 ± 1.26, p < 0.0001) was significantly increased as compared to those with compensated (3.25 ± 1.60). PDPN score positively and significantly correlated with the number of IELs (r = 0.33), serum TNF-α (r = 0.35), and IL-6 (r = 0.48) levels, while inversely correlated with TJP1 expression (r = -0.46, p < 0.05 each). In Cox regression, the PDPN score was a significant and independent 3-month-mortality predictor in patients (HR: 5.61; 1.08-29.109; p = 0.04). The area under the curve for the PDPN score was 84.2, and cutoff value for predicting mortality was ≥6.5 with 100% sensitivity and 75% specificity. Collectively, dilated LVs with high PDPN expression in D2-biopsies is a characteristic feature of patients with decompensated cirrhosis. PDPN score correlates with enhanced gut and systemic inflammation and also associates with 3-month mortality in cirrhosis.
Collapse
Affiliation(s)
- Pinky Juneja
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Aarti Sharma
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - S. M. Shasthry
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Guresh Kumar
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Dinesh M. Tripathi
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - V. Rajan
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Archana Rastogi
- Department of Pathology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Shiv K. Sarin
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Savneet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| |
Collapse
|
6
|
Pyropia yezoensis-derived porphyran attenuates acute and chronic colitis by suppressing dendritic cells. Int J Biol Macromol 2023; 231:123148. [PMID: 36639074 DOI: 10.1016/j.ijbiomac.2023.123148] [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: 09/19/2022] [Revised: 12/26/2022] [Accepted: 01/02/2023] [Indexed: 01/12/2023]
Abstract
Porphyran is known to inhibit immune cell function. Previously, porphyran was shown to prevent lipopolysaccharide-induced sepsis in mice. However, studies on the inhibitory effects of porphyran during colitis are currently lacking. In this study, we evaluated the effects of Pyropia yezoensis-derived porphyran on dextran sodium sulfate (DSS)-induced acute and chronic colitis. The oral or intraperitoneal administration of porphyran inhibited the progression of DSS-induced colitis in mice, with the former also preventing immune cell infiltration in the colon. The levels of intracellular interferon-γ and interleukin-17 in T cells decreased when porphyran was administered orally. Porphyran inhibited T cell activation by suppressing dendritic cells (DCs) and macrophages. Porphyran prevented pathogen-associated molecular pattern and damage-associated molecular pattern-dependent DC and macrophage activation. Finally, porphyran attenuated chronic colitis caused via the long-term administration of DSS. These findings indicate that the oral administration of porphyran can inhibit DSS-induced colitis by suppressing DC and macrophage activation.
Collapse
|
7
|
Abdelsalam HM, Helal MG, Abu-Elsaad NM. TLR4-IN-C34 protects against acute kidney injury via modulating TLR4/MyD88/NF-κb axis, MAPK, and apoptosis. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:1334-1340. [PMID: 36474570 PMCID: PMC9699956 DOI: 10.22038/ijbms.2022.67168.14727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 10/09/2022] [Indexed: 01/25/2023]
Abstract
OBJECTIVES Acute kidney injury (AKI) is a major component of isoproterenol (ISO) induced cardiorenal syndrome. In this study, we investigated the effect of TLR4-IN-C34 as a toll-like receptor (TLR)-4 inhibitor on ameliorating ISO-induced AKI and the possible molecular underlying pathways. MATERIALS AND METHODS The study included 4 groups: control group, ISO group (rats received 100 mg/kg ISO in 2 doses 24 hr apart, SC), ISO+C341 and ISO+C343 groups (rats received 1 or 3 mg/kg TLR4-IN-C34 respectively twice one hour before each ISO injection, IP). RESULTS Obtained results showed that TLR4-IN-C34 injection prior to ISO decreased serum creatinine level (P<0.05). Renal tissue histopathologic changes were markedly decreased by TLR4-IN-C34. Renal relative expression of MAPK and MyD88 mRNA decreased significantly in both ISO+C341 and ISO+C343 groups compared with the ISO group (P<0.05). Furthermore, TLR-IN-C34 lowered the inflammatory cytokines IL-8, IL-1β, and IL-12 renal levels (P<0.05). Immunostained kidney sections showed a marked decrease in NF-κb positive cells in addition to the apoptotic marker Bax (P<0.05) by the two tested doses of TLR4-IN-C34. On the other hand, the expression of the antiapoptotic marker Bcl-2 by renal cells was markedly increased. CONCLUSION It can be concluded that TLR4-IN-C34 ameliorates ISO-induced AKI through anti-inflammatory anti-apoptotic effects and modulation of TLR4 signaling pathways.
Collapse
Affiliation(s)
- Hadeer M. Abdelsalam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt, Department of Pharmacology, Faculty of Pharmacy, Horus University, Egypt
| | - Manar G. Helal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Nashwa M. Abu-Elsaad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt,Corresponding author: Nashwa M Abu-Elsaad. Pharmacology and Toxicology Dep. Faculty of Pharmacy, Mansoura University, El Gomhoria street, Eldakahlia, 35516, Egypt.
| |
Collapse
|
8
|
Stephens M. The emerging potential of Aptamers as therapeutic agents in infection and inflammation. Pharmacol Ther 2022; 238:108173. [DOI: 10.1016/j.pharmthera.2022.108173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 03/10/2022] [Accepted: 03/15/2022] [Indexed: 10/18/2022]
|
9
|
Lee HS, Kim EN, Jeong GS. Ameliorative Effect of Citropten Isolated from Citrusaurantifolia Peel Extract as a Modulator of T Cell and Intestinal Epithelial Cell Activity in DSS-Induced Colitis. Molecules 2022; 27:4633. [PMID: 35889507 PMCID: PMC9321940 DOI: 10.3390/molecules27144633] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/18/2022] [Accepted: 07/18/2022] [Indexed: 02/01/2023] Open
Abstract
Citropten is a coumarin that is mainly found in fruits of Rutaceae trees, but its anti-inflammatory activities in colitis is still unknown. In this study, we investigated its attenuating effect of citropten isolated from Citrus aurantifolia extract on DSS-induced colitis through the modulation of the activity of T cells and intestinal epithelial cells. We found that pre-treatment with citropten downregulates the activity of T cells and intestinal epithelial cells without a negative effect on the viability of Jurkat and HT-29 cells. The results from the Western blot analysis revealed that pre-treatment with citropten reduces the NFκB and MAPK signaling pathway in activated T cells and intestinal epithelial cells. We elucidated that the oral administration of citropten alleviates the colonic inflammation and activity of effector T cells in DSS-induced colitis by measuring changes in body weight, histological scoring from H&E-stained sections, mRNA levels of pro-inflammatory cytokines and the phosphorylation level of the MAPK signaling pathway.
Collapse
Affiliation(s)
- Hyun-Su Lee
- Department of Physiology, School of Medicine, Daegu Catholic University, Daegu 42472, Korea;
| | - Eun-Nam Kim
- College of Pharmacy, Chungnam National University, Daejeon 34134, Korea;
| | - Gil-Saeng Jeong
- College of Pharmacy, Chungnam National University, Daejeon 34134, Korea;
| |
Collapse
|
10
|
Stephens M, Keane K, Roizes S, Liao S, Weid PYVD. Mincle-binding DNA aptamer demonstrates therapeutic potential in a model of inflammatory bowel disease. MOLECULAR THERAPY - NUCLEIC ACIDS 2022; 28:935-947. [PMID: 35782912 PMCID: PMC9207717 DOI: 10.1016/j.omtn.2022.05.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 05/12/2022] [Indexed: 10/25/2022]
|
11
|
Herrada AA, Olate-Briones A, Lazo-Amador R, Liu C, Hernández-Rojas B, Riadi G, Escobedo N. Lymph Leakage Promotes Immunosuppression by Enhancing Anti-Inflammatory Macrophage Polarization. Front Immunol 2022; 13:841641. [PMID: 35663931 PMCID: PMC9160822 DOI: 10.3389/fimmu.2022.841641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 04/25/2022] [Indexed: 11/16/2022] Open
Abstract
Lymphatic vasculature is a network of capillaries and vessels capable of draining extracellular fluid back to blood circulation and to facilitate immune cell migration. Although the role of the lymphatic vasculature as coordinator of fluid homeostasis has been extensively studied, the consequences of abnormal lymphatic vasculature function and impaired lymph drainage have been mostly unexplored. Here, by using the Prox1+/- mice with defective lymphatic vasculature and lymphatic leakage, we provide evidence showing that lymph leakage induces an immunosuppressive environment by promoting anti-inflammatory M2 macrophage polarization in different inflammatory conditions. In fact, by using a mouse model of tail lymphedema where lymphatic vessels are thermal ablated leading to lymph accumulation, an increasing number of anti-inflammatory M2 macrophages are found in the lymphedematous tissue. Moreover, RNA-seq analysis from different human tumors shows that reduced lymphatic signature, a hallmark of lymphatic dysfunction, is associated with increased M2 and reduced M1 macrophage signatures, impacting the survival of the patients. In summary, we show that lymphatic vascular leakage promotes an immunosuppressive environment by enhancing anti-inflammatory macrophage differentiation, with relevance in clinical conditions such as inflammatory bowel diseases or cancer.
Collapse
Affiliation(s)
- Andrés A. Herrada
- Lymphatic Vasculature and Inflammation Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca, Chile
| | - Alexandra Olate-Briones
- Lymphatic Vasculature and Inflammation Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca, Chile
| | - Rodrigo Lazo-Amador
- Lymphatic Vasculature and Inflammation Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca, Chile
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bairon Hernández-Rojas
- Ph.D Program in Sciences Mention in Modeling of Chemical and Biological Systems, Faculty of Engineering, University of Talca, Talca, Chile
| | - Gonzalo Riadi
- Agencia Nacional de Investigación y Desarrollo (ANID) – Millennium Science Initiative Program Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Center for Bioinformatics, Simulation and Modeling, CBSM, Department of Bioinformatics, Faculty of Engineering, University of Talca, Talca, Chile
| | - Noelia Escobedo
- Lymphatic Vasculature and Inflammation Research Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca, Chile
| |
Collapse
|
12
|
Liu HY, Gu F, Zhu C, Yuan L, Zhu C, Zhu M, Yao J, Hu P, Zhang Y, Dicksved J, Bao W, Cai D. Epithelial Heat Shock Proteins Mediate the Protective Effects of Limosilactobacillus reuteri in Dextran Sulfate Sodium-Induced Colitis. Front Immunol 2022; 13:865982. [PMID: 35320932 PMCID: PMC8934773 DOI: 10.3389/fimmu.2022.865982] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 02/11/2022] [Indexed: 11/13/2022] Open
Abstract
Defects in gut barrier function are implicated in gastrointestinal (GI) disorders like inflammatory bowel disease (IBD), as well as in systemic inflammation. With the increasing incidence of IBD worldwide, more attention should be paid to dietary interventions and therapeutics with the potential to boost the natural defense mechanisms of gut epithelial cells. The current study aimed to investigate the protective effects of Limosilactobacillus reuteri ATCC PTA 4659 in a colitis mouse model and delineate the mechanisms behind it. Wild-type mice were allocated to the control group; or given 3% dextran sulfate sodium (DSS) in drinking water for 7 days to induce colitis; or administered L. reuteri for 7 days as pretreatment; or for 14 days starting 7 days before subjecting to the DSS. Peroral treatment with L. reuteri improved colitis severity clinically and morphologically and reduced the colonic levels of Tumor necrosis factor-α (TNF-α) (Tnf), Interleukin 1-β (Il1β), and nterferon-γ (Ifng), the crucial pro-inflammatory cytokines in colitis onset. It also prevented the CD11b+Ly6G+ neutrophil recruitment and the skewed immune responses in mesenteric lymph nodes (MLNs) of CD11b+CD11c+ dendritic cell (DC) expansion and Foxp3+CD4+ T-cell reduction. Using 16S rRNA gene amplicon sequencing and RT-qPCR, we demonstrated a colitis-driven bacterial translocation to MLNs and gut microbiota dysbiosis that were in part counterbalanced by L. reuteri treatment. Moreover, the expression of barrier-preserving tight junction (TJ) proteins and cytoprotective heat shock protein (HSP) 70 and HSP25 was reduced by colitis but boosted by L. reuteri treatment. A shift in expression pattern was also observed with HSP70 in response to the pretreatment and with HSP25 in response to L. reuteri-DSS. In addition, the changes of HSPs were found to be correlated to bacterial load and epithelial cell proliferation. In conclusion, our results demonstrate that the human-derived L. reuteri strain 4659 confers protection in experimental colitis in young mice, while intestinal HSPs may mediate the probiotic effects by providing a supportive protein–protein network for the epithelium in health and colitis.
Collapse
Affiliation(s)
- Hao-Yu Liu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Fang Gu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Cuipeng Zhu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Long Yuan
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Chuyang Zhu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Miaonan Zhu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Jiacheng Yao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Ping Hu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Yunzeng Zhang
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Johan Dicksved
- Department of Animal Nutrition and Management, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Wenbin Bao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- *Correspondence: Demin Cai, ; Wenbin Bao,
| | - Demin Cai
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- *Correspondence: Demin Cai, ; Wenbin Bao,
| |
Collapse
|
13
|
Jo M, Lee J, Kim HG, Kim JK, Kim H, Shin KK, Bach TT, Eum SM, Lee JS, Choung ES, Yang Y, Kim KH, Sung GH, Yoo BC, Cho JY. Anti-inflammatory effect of Barringtonia angusta methanol extract is mediated by targeting of Src in the NF-κB signalling pathway. PHARMACEUTICAL BIOLOGY 2021; 59:799-810. [PMID: 34190667 PMCID: PMC8253214 DOI: 10.1080/13880209.2021.1938613] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 04/27/2021] [Accepted: 05/28/2021] [Indexed: 06/13/2023]
Abstract
CONTEXT Among the plants in the genus Barringtonia (Lecythidaceae) used as traditional medicines to treat arthralgia, chest pain, and haemorrhoids in Indonesia, Barringtonia racemosa L. and Barringtonia acutangula (L.) Gaertn. have demonstrated anti-inflammatory activity in systemic inflammatory models. OBJECTIVE The anti-inflammatory activity of Barringtonia angusta Kurz has not been investigated. We prepared a methanol extract of the leaves and stems of B. angusta (Ba-ME) and systemically evaluated its anti-inflammatory effects in vitro and in vivo. MATERIALS AND METHODS RAW264.7 cells stimulated with LPS or Pam3CSK4 for 24 h were treated with Ba-ME (12.5, 25, 50, 100, and 150 µg/mL), and NO production and mRNA levels of inflammatory genes were evaluated. Luciferase reporter gene assay, western blot analysis, overexpression experiments, and cellular thermal shift assay were conducted to explore the mechanism of Ba-ME. In addition, the anti-gastritis activity of Ba-ME (50 and 100 mg/kg, administered twice per day for two days) was evaluated using an HCl/EtOH-induced gastritis mouse model. RESULTS Ba-ME dose-dependently suppressed NO production [IC50 = 123.33 µg/mL (LPS) and 46.89 µg/mL (Pam3CSK4)] without affecting cell viability. Transcriptional expression of iNOS, IL-1β, COX-2, IL-6, and TNF-α and phosphorylation of Src, IκBα, p50/105, and p65 were inhibited by Ba-ME. The extract specifically targeted the Src protein by binding to its SH2 domain. Moreover, Ba-ME significantly ameliorated inflammatory lesions in the HCl/EtOH-induced gastritis model. DISCUSSION AND CONCLUSIONS The anti-inflammatory activity of Ba-ME is mediated by targeting of the Src/NF-κB signalling pathway, and B. angusta has potential as an anti-inflammatory drug.
Collapse
Affiliation(s)
- Minkyeong Jo
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jongsung Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
- Research Institute of Biomolecule Control and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Republic of Korea
| | - Han Gyung Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
- Research Institute of Biomolecule Control and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Republic of Korea
| | - Jin Kyeong Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Haeyeop Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Kon Kuk Shin
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Tran The Bach
- Institute of Ecology and Biological Resources, Vietnam Academy of Science and Technology (VAST), Ha Noi, Vietnam
| | - Sang Mi Eum
- International Biological Material Research Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon, Republic of Korea
| | | | | | - Yoonyong Yang
- Biological and Genetic Resources Assessment Division, National Institute of Biological Resources, Incheon, Republic of Korea
| | - Kyung-Hee Kim
- Proteomic Analysis Team, Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Gi-Ho Sung
- Department of Microbiology, Biomedical Institute of Mycological Resource, International St. Mary's Hospital and College of Medicine, Catholic Kwandong University, Incheon, Republic of Korea
| | - Byong Chul Yoo
- Division of Translational Science, Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
- Research Institute of Biomolecule Control and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
14
|
Liu XH, Wu LM, Wang JL, Dong XH, Zhang SC, Li XH, Xu H, Liu DB, Li ZH, Liu ZM, Wu SG, Hu YW. Long non-coding RNA RP11-490M8.1 inhibits lipopolysaccharide-induced pyroptosis of human umbilical vein endothelial cells via the TLR4/NF-κB pathway. Immunobiology 2021; 226:152133. [PMID: 34469785 DOI: 10.1016/j.imbio.2021.152133] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 08/03/2021] [Accepted: 08/23/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Pyroptosis is a relatively newly discovered form of programmed cell death that plays an important role in the development of atherosclerosis. Many studies have reported that lncRNAs participated in the regulation of atherosclerosis development. However, the regulatory mechanism of lncRNAs in pyroptosis must be studied further. METHODS In a previous study, microarray analysis was used to detect the lncRNA expression profile in three human advanced atherosclerotic plaques and three normal arterial intimae. In the present research, in vitro assays were performed to investigate the role of lncRNA RP11-490M8.1 on pyroptosis. The relative gene mRNA and lncRNA expression levels were tested by quantitative real-time PCR, and protein levels were evaluated by western blot analysis. The RNA hybrid structure was analyzed using the DINAMelt server. RESULTS The lncRNA RP11-490M8.1 was significantly downregulated in atherosclerotic plaques and serum. Lipopolysaccharide (LPS) markedly reduced the expression of lncRNA RP11-490M8.1 and induced pyroptosis by increasingthe mRNA and protein levels of NLRP3, caspase-1, ASC, IL-1β, and IL-18 in HUVECs. The promotion effects ofLPS on pyroptosis were markedly suppressed by overexpression of lncRNA RP11-490M8.1. In addition, LPS increased the mRNA and protein levels ofTLR4 and NF-κB, which was also markedly offsetby overexpression of lncRNA RP11-490M8.1. CONCLUSIONS These findings indicated that lncRNA RP11-490M8.1 inhibited LPS-induced pyroptosis via the TLR4/NF-κB pathway. Thus, lncRNA RP11-490M8.1 may provide a therapeutic target to ameliorate atherosclerosis.
Collapse
Affiliation(s)
- Xue-Hui Liu
- Department of Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China; Department of Clinical Laboratory, Guangzhou Twelfth People's Hospital, Guangzhou, Guangdong 510620, China
| | - Li-Mei Wu
- Department of Clinical Laboratory, Guangzhou Twelfth People's Hospital, Guangzhou, Guangdong 510620, China
| | - Jia-Li Wang
- Department of Blood Transfusion, Linyi People's Hospital of Shandong Province, Linyi, ShanDong 276000, China
| | - Xian-Hui Dong
- Department of Clinical Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong 510623, China
| | - Shun-Chi Zhang
- Department of Clinical Laboratory, Guangzhou Twelfth People's Hospital, Guangzhou, Guangdong 510620, China
| | - Xue-Heng Li
- Department of Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Hui Xu
- Traditional Chinese Medical Hospital of Qingyuan, Qingyuan, Guangdong 511500, China
| | - Da-Bin Liu
- Department of Clinical Laboratory, Guangzhou Twelfth People's Hospital, Guangzhou, Guangdong 510620, China
| | - Zhi-Hai Li
- Department of Clinical Laboratory, Guangzhou Twelfth People's Hospital, Guangzhou, Guangdong 510620, China
| | - Zhe-Ming Liu
- Stomatology Major, Medical College of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Shao-Guo Wu
- Department of Clinical Laboratory, Guangzhou Twelfth People's Hospital, Guangzhou, Guangdong 510620, China.
| | - Yan-Wei Hu
- Department of Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China; Department of Clinical Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong 510623, China.
| |
Collapse
|
15
|
Shaikh H, Vargas JG, Mokhtari Z, Jarick KJ, Ulbrich M, Mosca JP, Viera EA, Graf C, Le DD, Heinze KG, Büttner-Herold M, Rosenwald A, Pezoldt J, Huehn J, Beilhack A. Mesenteric Lymph Node Transplantation in Mice to Study Immune Responses of the Gastrointestinal Tract. Front Immunol 2021; 12:689896. [PMID: 34381447 PMCID: PMC8352558 DOI: 10.3389/fimmu.2021.689896] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/08/2021] [Indexed: 02/02/2023] Open
Abstract
Mesenteric lymph nodes (mLNs) are sentinel sites of enteral immunosurveillance and immune homeostasis. Immune cells from the gastrointestinal tract (GIT) are constantly recruited to the mLNs in steady-state and under inflammatory conditions resulting in the induction of tolerance and immune cells activation, respectively. Surgical dissection and transplantation of lymph nodes (LN) is a technique that has supported seminal work to study LN function and is useful to investigate resident stromal and endothelial cell biology and their cellular interactions in experimental disease models. Here, we provide a detailed protocol of syngeneic mLN transplantation and report assays to analyze effective mLN engraftment in congenic recipients. Transplanted mLNs allow to study T cell activation and proliferation in preclinical mouse models. Donor mLNs proved viable and functional after surgical transplantation and regenerated blood and lymphatic vessels. Immune cells from the host completely colonized the transplanted mLNs within 7-8 weeks after the surgical intervention. After allogeneic hematopoietic cell transplantation (allo-HCT), adoptively transferred allogeneic CD4+ T cells from FVB/N (H-2q) mice homed to the transplanted mLNs in C57BL/6 (H-2b) recipients during the initiation phase of acute graft-versus-host disease (aGvHD). These CD4+ T cells retained full proliferative capacity and upregulated effector and gut homing molecules comparable to those in mLNs from unmanipulated wild-type recipients. Wild type mLNs transplanted into MHCII deficient syngeneic hosts sufficed to activate alloreactive T cells upon allogeneic hematopoietic cell transplantation, even in the absence of MHCII+ CD11c+ myeloid cells. These data support that orthotopically transplanted mLNs maintain physiological functions after transplantation. The technique of LN transplantation can be applied to study migratory and resident cell compartment interactions in mLNs as well as immune reactions from and to the gut under inflammatory and non-inflammatory conditions.
Collapse
Affiliation(s)
- Haroon Shaikh
- Interdisciplinary Center for Clinical Research (IZKF) Experimental Stem Cell Transplantation Laboratory, Würzburg University Hospital, Würzburg, Germany
- Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
- Graduate School of Life Sciences, Würzburg University, Würzburg, Germany
| | - Juan Gamboa Vargas
- Interdisciplinary Center for Clinical Research (IZKF) Experimental Stem Cell Transplantation Laboratory, Würzburg University Hospital, Würzburg, Germany
- Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
- Graduate School of Life Sciences, Würzburg University, Würzburg, Germany
| | - Zeinab Mokhtari
- Interdisciplinary Center for Clinical Research (IZKF) Experimental Stem Cell Transplantation Laboratory, Würzburg University Hospital, Würzburg, Germany
- Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
| | - Katja J. Jarick
- Interdisciplinary Center for Clinical Research (IZKF) Experimental Stem Cell Transplantation Laboratory, Würzburg University Hospital, Würzburg, Germany
- Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
- Graduate School of Life Sciences, Würzburg University, Würzburg, Germany
| | - Maria Ulbrich
- Interdisciplinary Center for Clinical Research (IZKF) Experimental Stem Cell Transplantation Laboratory, Würzburg University Hospital, Würzburg, Germany
- Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
| | - Josefina Peña Mosca
- Interdisciplinary Center for Clinical Research (IZKF) Experimental Stem Cell Transplantation Laboratory, Würzburg University Hospital, Würzburg, Germany
- Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
- Graduate School of Life Sciences, Würzburg University, Würzburg, Germany
| | - Estibaliz Arellano Viera
- Interdisciplinary Center for Clinical Research (IZKF) Experimental Stem Cell Transplantation Laboratory, Würzburg University Hospital, Würzburg, Germany
- Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
| | - Caroline Graf
- Interdisciplinary Center for Clinical Research (IZKF) Experimental Stem Cell Transplantation Laboratory, Würzburg University Hospital, Würzburg, Germany
- Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
| | - Duc-Dung Le
- Interdisciplinary Center for Clinical Research (IZKF) Experimental Stem Cell Transplantation Laboratory, Würzburg University Hospital, Würzburg, Germany
- Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
| | - Katrin G. Heinze
- Graduate School of Life Sciences, Würzburg University, Würzburg, Germany
- Rudolf Virchow Center, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Maike Büttner-Herold
- Department of Nephropathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Andreas Rosenwald
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
- Comprehensive Cancer Centre Mainfranken, Würzburg University Hospital, Würzburg, Germany
| | - Joern Pezoldt
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Jochen Huehn
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Andreas Beilhack
- Interdisciplinary Center for Clinical Research (IZKF) Experimental Stem Cell Transplantation Laboratory, Würzburg University Hospital, Würzburg, Germany
- Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
- Graduate School of Life Sciences, Würzburg University, Würzburg, Germany
| |
Collapse
|
16
|
Mollace A, Coluccio ML, Donato G, Mollace V, Malara N. Cross-talks in colon cancer between RAGE/AGEs axis and inflammation/immunotherapy. Oncotarget 2021; 12:1281-1295. [PMID: 34194625 PMCID: PMC8238251 DOI: 10.18632/oncotarget.27990] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/02/2021] [Indexed: 12/19/2022] Open
Abstract
The tumour microenvironment is the result of the activity of many types of cells in various metabolic states, whose metabolites are shared between cells. This cellular complexity results in an availability profile of nutrients and reactive metabolites such as advanced glycation end products (AGE). The tumour microenvironment is not favourable to immune cells due to hypoxia and for the existence of significant competition between various types of cells for a limited nutrient pool. However, it is now known that cancer cells can influence the host's immune reaction through the expression and secretion of numerous molecules. The microenvironment can therefore present itself in different patterns that contribute to shaping immune surveillance. Colorectal cancer (CRC) is one of the most important causes of death in cancer patients. Recently, immunotherapy has begun to give encouraging results in some groups of patients suffering from this neoplasm. The analysis of literature data shows that the RAGE (Receptor for advanced glycation end products) and its numerous ligands contribute to connect the energy metabolic pathway, which appears prevalently disconnected by mitochondrial running, with the immune reaction, conditioned by local microbiota and influencing tumour growth. Understanding how metabolism in cancer and immune cells shapes response and resistance to therapy, will provide novel potential strategies to increase both the number of tumour types treated by immunotherapy and the rate of immunotherapy response. The analysis of literature data shows that an immunotherapy approach based on the knowledge of RAGE and its ligands is not only possible, but also desirable in the treatment of CRC.
Collapse
Affiliation(s)
- Annachiara Mollace
- Department of Health Sciences, Research Centre IRC-FSH, University Magna Græcia of Catanzaro, 88100 Catanzaro, Italy
| | - Maria Laura Coluccio
- Department of Experimental and Clinical Medicine, Bionem Laboratory, Magna Græcia University of Catanzaro, 88100 Catanzaro, Italy
| | - Giuseppe Donato
- Department of Health Sciences, University Magna Græcia of Catanzaro, Campus S. Venuta, 88100 Catanzaro, Italy
| | - Vincenzo Mollace
- Department of Health Sciences, Research Centre IRC-FSH, University Magna Græcia of Catanzaro, 88100 Catanzaro, Italy.,These authors contributed equally to this work
| | - Natalia Malara
- Department of Experimental and Clinical Medicine, Bionem Laboratory, Magna Græcia University of Catanzaro, 88100 Catanzaro, Italy.,These authors contributed equally to this work
| |
Collapse
|
17
|
Ocansey DKW, Pei B, Xu X, Zhang L, Olovo CV, Mao F. Cellular and molecular mediators of lymphangiogenesis in inflammatory bowel disease. J Transl Med 2021; 19:254. [PMID: 34112196 PMCID: PMC8190852 DOI: 10.1186/s12967-021-02922-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/02/2021] [Indexed: 02/07/2023] Open
Abstract
Background Recent studies reporting the intricate crosstalk between cellular and molecular mediators and the lymphatic endothelium in the development of inflammatory bowel diseases (IBD) suggest altered inflammatory cell drainage and lymphatic vasculature, implicating the lymphatic system as a player in the occurrence, development, and recurrence of intestinal diseases. This article aims to review recent data on the modulatory functions of cellular and molecular components of the IBD microenvironment on the lymphatic system, particularly lymphangiogenesis. It serves as a promising therapeutic target for IBD management and treatment. The interaction with gut microbiota is also explored. Main text Evidence shows that cells of the innate and adaptive immune system and certain non-immune cells participate in the complex processes of inflammatory-induced lymphangiogenesis through the secretion of a wide spectrum of molecular factors, which vary greatly among the various cells. Lymphangiogenesis enhances lymphatic fluid drainage, hence reduced infiltration of immunomodulatory cells and associated-inflammatory cytokines. Interestingly, some of the cellular mediators, including mast cells, neutrophils, basophils, monocytes, and lymphatic endothelial cells (LECs), are a source of lymphangiogenic molecules, and a target as they express specific receptors for lymphangiogenic factors. Conclusion The effective target of lymphangiogenesis is expected to provide novel therapeutic interventions for intestinal inflammatory conditions, including IBD, through both immune and non-immune cells and based on cellular and molecular mechanisms of lymphangiogenesis that facilitate inflammation resolution.
Collapse
Affiliation(s)
- Dickson Kofi Wiredu Ocansey
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China.,Directorate of University Health Services, University of Cape Coast, Cape Coast, Ghana
| | - Bing Pei
- Department of Clinical Laboratory, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, 223800, Jiangsu, People's Republic of China
| | - Xinwei Xu
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Lu Zhang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Chinasa Valerie Olovo
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China.,Department of Microbiology, University of Nigeria, Nsukka, 410001, Nigeria
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China.
| |
Collapse
|
18
|
Circular RNA circHIPK3 Activates Macrophage NLRP3 Inflammasome and TLR4 Pathway in Gouty Arthritis via Sponging miR-561 and miR-192. Inflammation 2021; 44:2065-2077. [PMID: 34085163 DOI: 10.1007/s10753-021-01483-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 04/22/2021] [Accepted: 05/19/2021] [Indexed: 10/21/2022]
Abstract
Increasing evidences indicate that circular RNAs (circRNAs) play important roles in regulating gene expressions in various diseases. However, the role of circRNAs in inflammatory response of gouty arthritis remains unknown. This study aims to investigate the role and underlying mechanism of circHIPK3 in inflammatory response of gouty arthritis. Quantitative real-time PCR was used to detect the expressions of circHIPK3, miR-192 and miR-561. Western blot was used to detect the protein levels of TLR4, NLRP3, nuclear factor-κB (NF-κB) related proteins, and Caspase-1. Dual luciferase reporter assay, RNA pull-down assay, and FISH assay were used to confirm the interaction between circHIPK3 and miR-192/miR-561. ELISA was used to detect interleukin (IL)-1β and tumor necrosis factor (TNF)-α levels. circHIPK3 was elevated in synovial fluid mononuclear cells (SFMCs) from patients with gouty arthritis and monosodium urate (MSU)-stimulated THP-1 cells. circHIPK3 overexpression promoted the inflammatory cytokines levels in MSU-stimulated THP-1 cells, and circHIPK3 silencing obtained the opposite effect. Mechanistically, circHIPK3 sponged miR-192 and miR-561, and subsequently promoted the expressions of miR-192 and miR-561 target gene TLR4 and NLRP3. In vivo experiments confirmed circHIPK3 knockdown suppressed gouty arthritis. circHIPK3 sponges miR-192 and miR-561 to promote TLR4 and NLRP3 expressions, thereby promoting inflammatory response in gouty arthritis.
Collapse
|
19
|
Zhang L, Ocansey DKW, Liu L, Olovo CV, Zhang X, Qian H, Xu W, Mao F. Implications of lymphatic alterations in the pathogenesis and treatment of inflammatory bowel disease. Biomed Pharmacother 2021; 140:111752. [PMID: 34044275 DOI: 10.1016/j.biopha.2021.111752] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/06/2021] [Accepted: 05/19/2021] [Indexed: 02/07/2023] Open
Abstract
Inflammatory bowel disease (IBD) is characterized by intense immune dysregulation, gut microbiota imbalance, and intestinal epithelium destruction. Among the factors that contribute to the pathogenesis of IBD, lymphatics have received less attention, hence less studied, characterized, and explored. However, in recent years, the role of the lymphatic system in gastrointestinal pathophysiology continues to be highlighted. This paper examines the implications of lymphatic changes in IBD pathogenesis related to immune cells, gut microbiota, intestinal and mesenteric epithelial barrier integrity, and progression to colorectal cancer (CRC). Therapeutic targets of lymphatics in IBD studies are also presented. Available studies indicate that lymph nodes and other secondary lymphatic tissues, provide highly specialized microenvironments for mounting effective immune responses and that lymphatic integrity plays a significant role in small intestine homeostasis, where the lymphatic vasculature effectively controls tissue edema, leukocyte exit, bacterial antigen, and inflammatory chemokine clearance. In IBD, there are functional and morphological alterations in intestinal and mesenteric lymphatic vessels (more profoundly in Crohn's disease [CD] compared to ulcerative colitis [UC]), including lymphangiogenesis, lymphangiectasia, lymphadenopathy, and lymphatic vasculature blockade, affecting not only immunity but gut microbiota and epithelial barrier integrity. While increased lymphangiogenesis is primarily associated with a good prognosis of IBD, increased lymphangiectasia, lymphadenopathy, and lymphatic vessel occlusion correlate with poor prognosis. IBD therapies that target the lymphatic system seek to increase lymphangiogenesis via induction of lymphangiogenic factors and inhibition of its antagonists. The resultant increased lymphatic flow coupled with other anti-inflammatory activities restores gut homeostasis.
Collapse
Affiliation(s)
- Lu Zhang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Dickson Kofi Wiredu Ocansey
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China; Directorate of University Health Services, University of Cape Coast, Cape Coast, Ghana
| | - Lianqin Liu
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Chinasa Valerie Olovo
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China; Department of Microbiology, University of Nigeria, Nsukka 410001, Nigeria
| | - Xu Zhang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Hui Qian
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Wenrong Xu
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China.
| |
Collapse
|
20
|
Impact of gastrointestinal tract variability on oral drug absorption and pharmacokinetics: An UNGAP review. Eur J Pharm Sci 2021; 162:105812. [PMID: 33753215 DOI: 10.1016/j.ejps.2021.105812] [Citation(s) in RCA: 140] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 02/19/2021] [Accepted: 03/16/2021] [Indexed: 12/17/2022]
Abstract
The absorption of oral drugs is frequently plagued by significant variability with potentially serious therapeutic consequences. The source of variability can be traced back to interindividual variability in physiology, differences in special populations (age- and disease-dependent), drug and formulation properties, or food-drug interactions. Clinical evidence for the impact of some of these factors on drug pharmacokinetic variability is mounting: e.g. gastric pH and emptying time, small intestinal fluid properties, differences in pediatrics and the elderly, and surgical changes in gastrointestinal anatomy. However, the link of colonic factors variability (transit time, fluid composition, microbiome), sex differences (male vs. female) and gut-related diseases (chronic constipation, anorexia and cachexia) to drug absorption variability has not been firmly established yet. At the same time, a way to decrease oral drug pharmacokinetic variability is provided by the pharmaceutical industry: clinical evidence suggests that formulation approaches employed during drug development can decrease the variability in oral exposure. This review outlines the main drivers of oral drug exposure variability and potential approaches to overcome them, while highlighting existing knowledge gaps and guiding future studies in this area.
Collapse
|
21
|
Stephens M, Liao S, von der Weid PY. Ultra-purification of Lipopolysaccharides reveals species-specific signalling bias of TLR4: importance in macrophage function. Sci Rep 2021; 11:1335. [PMID: 33446670 PMCID: PMC7809447 DOI: 10.1038/s41598-020-79145-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 11/30/2020] [Indexed: 01/03/2023] Open
Abstract
TLR4 location, and bacterial species-derived lipopolysaccharides, play a significant role in the downstream activation of transcription factors, accessory molecules, and products. Here, this is demonstrated through the use of classically-activated and alternatively-activated macrophages. We show that, when polarized, human macrophages differentially express and localize TLR4, resulting in biased recognition and subsequent signalling of LPS derived from Pseudomonas aeruginosa, Escherichia coli, and Salmonella enterica. Analysis of activation demonstrated that in classically activated macrophages, P. aeruginosa signals from the plasma membrane via TLR4 to p65 dependent on TAK1 and TBK1 signalling. E. coli signals dependent or independent of the endosome, utilizing both TAK1- and TBK1-signalling to induce P65 and IRF3 inducible genes and cytokines. S. enterica however, only induces P65 and IRF3 phosphorylation through signalling via the endosome. This finding outlines clear signalling mechanisms by which innate immune cells, such as macrophages, can distinguish between bacterial species and initiate specialized responses through TLR4.
Collapse
Affiliation(s)
- Matthew Stephens
- Department of Physiology and Pharmacology, Inflammation Research Network, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Shan Liao
- Department of Microbiology, Immunology & Infectious Disease, Inflammation Research Network, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Pierre-Yves von der Weid
- Department of Physiology and Pharmacology, Inflammation Research Network, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Alberta, Canada.
| |
Collapse
|
22
|
Xu Z, Lin R, Hou X, Wu J, Zhao W, Ma H, Fan Z, Li S, Zhu Y, Zhang D. Immunomodulatory mechanism of a purified polysaccharide isolated from Isaria cicadae Miquel on RAW264.7 cells via activating TLR4-MAPK-NF-κB signaling pathway. Int J Biol Macromol 2020; 164:4329-4338. [DOI: 10.1016/j.ijbiomac.2020.09.035] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 05/29/2020] [Accepted: 09/05/2020] [Indexed: 12/19/2022]
|
23
|
Wang R, Zhang M, Hu S, Liu K, Tai Y, Tao J, Zhou W, Zhao Z, Wang Q, Wei W. Ginsenoside metabolite compound-K regulates macrophage function through inhibition of β-arrestin2. Biomed Pharmacother 2019; 115:108909. [PMID: 31071508 DOI: 10.1016/j.biopha.2019.108909] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/09/2019] [Accepted: 04/22/2019] [Indexed: 12/14/2022] Open
Abstract
Ginsenoside metabolite compound-K (C-K), which is an active metabolite of ginsenoside in vivo, can produce anti-inflammatory affects by activating glucocorticoid receptors (GRs) to inhibit the expression of β-arrestin2. Studies have shown that C-K can inhibit the function of immune cells including macrophage polarization and phagocytosis. However, the mechanism by which C-K regulates macrophage polarization is currently unclear. Toll-like receptors (TLRs) are the pattern recognition receptors on the membrane of immune cells, with TLR4 being especially important in polarization of macrophages. The Gαi-mediated activation of nuclear factor-κB (NF-κB) by TLR4 promotes inflammation and phagocytosis in macrophages by increasing the proportion of type I phenotypic macrophages (M1). Whether C-K inhibits the signal transduction of TLR4-Gαi-NF-κB and how that effects macrophage polarization regulation in murine models of RA is not reported. The coupling of G proteins with receptors is regulated by β-arrestin2, but it has been unclear whether C-K modulates the TLR4 interaction with G proteins by inhibiting the expression of β-arrestin2. To explore these questions, the collagen-induced arthritis (CIA) mouse model was employed, and mice were treated with C-K (112 mg/kg/day). The results depict that C-K treatment inhibits macrophage phagocytosis and reduces the proportion of M1. C-K decreases the overexpressed β-arrestin2, Gαi, TLR4 and NF-κB in macrophages of CIA mice, while increasing the expression of Gαs. Furthermore, C-K promotes TLR4-Gαs coupling and inhibits TLR4-Gαi coupling through β-arrestin2 regulation in macrophages, leading to a decrease in the proportion of M1 to M2 macrophages and improved outcomes in CIA mice.
Collapse
Affiliation(s)
- Rui Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology of Education, Ministry of China, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China
| | - Mei Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology of Education, Ministry of China, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China
| | - Shanshan Hu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology of Education, Ministry of China, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China
| | - Kangkang Liu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology of Education, Ministry of China, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China
| | - Yu Tai
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology of Education, Ministry of China, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China
| | - Juan Tao
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology of Education, Ministry of China, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China
| | - Weijie Zhou
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology of Education, Ministry of China, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China
| | - Zongbiao Zhao
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology of Education, Ministry of China, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China
| | - Qingtong Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology of Education, Ministry of China, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China.
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology of Education, Ministry of China, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, China.
| |
Collapse
|