1
|
He M, Lim XY, Li J, Li L, Zhang T. Mechanisms of acupuncture at Zusanli (ST36) and its combinational acupoints for stress gastric ulcer based on the correlation between Zang-fu and acupoints. JOURNAL OF INTEGRATIVE MEDICINE 2025; 23:1-11. [PMID: 39736482 DOI: 10.1016/j.joim.2024.12.003] [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: 09/17/2023] [Accepted: 08/12/2024] [Indexed: 01/01/2025]
Abstract
Gastric ulcer (GU) is a common digestive system disease. Acupuncture, as one of the external treatments of traditional Chinese medicine (TCM), has the characteristics of multi-target, multi-pathway and multi-level action in the treatment of GU. The relationship between meridian points and Zang-fu is an important part of the theory of TCM, which is crucial for the diagnosis and treatment of diseases. There is an external and internal link between acupoints and Zang-fu. The pathological reaction of Zang-fu can manifest as acupoint sensitization, while stimulation of acupoints can play a therapeutic role in the internal Zang-fu. Therefore, the acupoint has the functions of reflecting and treating diseases. This review explores the tender points on the body surface of patients with GU and the rules of acupoint selection. In addition, Zusanli (ST36), as one of the most used acupoints of the stomach meridian, was selected to show the mechanisms behind acupoint stimulation in the treatment of GU in greater detail, specifically in the well-studied model of the stress GU (SGU). Hence, the mechanisms of acupuncture at ST36 and points commonly used in combination with ST36 to treat SGU are discussed further. Treatment effects can be achieved through anti-inflammatory and antioxidant activities, gastric mucosal injury repair, and interaction with the brain-gut axis. In summary, this review provides evidence for a comprehensive understanding of the phenomena and mechanism of acupoint functions for GU. Please cite this article as: He M, Lim XY, Li J, Li L, Zhang T. Mechanisms of acupuncture at Zusanli (ST36) and its combinational acupoints for stress gastric ulcer based on the correlation between Zang-fu and acupoints. J Integr Med. 2025; 23(1): 1-11.
Collapse
Affiliation(s)
- Mu He
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xue Yee Lim
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jing Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ling Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
2
|
Kamianowska M, Rybi-Szumińska A, Kamianowska A, Maciejczyk M, Sołomianko K, Koput A, Wasilewska A. The Urinary Concentration of Trefoil Factor 3 (TFF3) in the Term and Preterm Neonates. J Clin Med 2023; 12:4936. [PMID: 37568337 PMCID: PMC10419516 DOI: 10.3390/jcm12154936] [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: 05/29/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND Distinguishing between a pathologic state and renal development is important in neonatology. Because the assessment of serum creatinine in neonates is not reliable, better biomarkers are needed. Trefoil factor 3 (TFF3) is proposed as a biomarker of kidney injury. The study aimed to assess its urinary concentration in healthy term and stable preterm neonates. MATERIAL AND METHODS The study included 80 term and 20 preterm neonates born in the Department of Perinatology of the University Clinical Hospital in Bialystok. Urine was obtained from the term neonates on the 1st day of life and from the preterm neonates on the 1st, 8th, 15th and 22nd day of life. The urinary concentration of TFF3 was determined using a commercially available immunoassay and was normalized for the urinary creatinine concentration (cr.). RESULTS The values of TFF3/cr. were higher in the preterm than in the term neonates (p < 0.05) (median (Q1-Q3): 1486.85 (614.92-3559.18) and 317.29 (68.07-671.40) ng/mg cr.). They did not differ in the subsequent days of the preterm neonates' lives. The ROC curve for TFF3/cr. in the preterm and term neonates showed AUC = 0.751 (cut-off value = 1684.25 ng/mg cr.). CONCLUSIONS Prematurity is associated with higher urinary excretion of TFF3. Male gender is associated with an increased urinary TFF3 excretion in term neonates.
Collapse
Affiliation(s)
- Monika Kamianowska
- Department of Neonatology and Neonatal Intensive Care, Medical University of Bialystok, M. C. Sklodowskiej 24a Street, 15-276 Białystok, Poland;
| | - Agnieszka Rybi-Szumińska
- Department of Pediatrics and Nephrology, Medical University of Bialystok, 15-269 Bialystok, Poland (A.K.)
| | - Aleksandra Kamianowska
- Department of Pediatrics and Nephrology, Medical University of Bialystok, 15-269 Bialystok, Poland (A.K.)
| | - Mateusz Maciejczyk
- Department of Hygiene, Epidemiology and Ergonomic, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Katarzyna Sołomianko
- Department of Neonatology and Neonatal Intensive Care, Medical University of Bialystok, M. C. Sklodowskiej 24a Street, 15-276 Białystok, Poland;
| | - Alicja Koput
- Department of Pediatric Laboratory Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Anna Wasilewska
- Department of Pediatrics and Nephrology, Medical University of Bialystok, 15-269 Bialystok, Poland (A.K.)
| |
Collapse
|
3
|
Yang Y, Lin Z, Lin Q, Bei W, Guo J. Pathological and therapeutic roles of bioactive peptide trefoil factor 3 in diverse diseases: recent progress and perspective. Cell Death Dis 2022; 13:62. [PMID: 35039476 PMCID: PMC8763889 DOI: 10.1038/s41419-022-04504-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 12/23/2021] [Accepted: 12/30/2021] [Indexed: 12/16/2022]
Abstract
Trefoil factor 3 (TFF3) is the last small-molecule peptide found in the trefoil factor family, which is mainly secreted by intestinal goblet cells and exerts mucosal repair effect in the gastrointestinal tract. Emerging evidence indicated that the TFF3 expression profile and biological effects changed significantly in pathological states such as cancer, colitis, gastric ulcer, diabetes mellitus, non-alcoholic fatty liver disease, and nervous system disease. More importantly, mucosal protection would no longer be the only effect of TFF3, it gradually exhibits carcinogenic activity and potential regulatory effect of nervous and endocrine systems, but the inner mechanisms remain unclear. Understanding the molecular function of TFF3 in specific diseases might provide a new insight for the clinical development of novel therapeutic strategies. This review provides an up-to-date overview of the pathological effects of TFF3 in different disease and discusses the binding proteins, signaling pathways, and clinical application.
Collapse
Affiliation(s)
- Yiqi Yang
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega, Guangzhou, China
| | - Ziyang Lin
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega, Guangzhou, China
| | - Quanyou Lin
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega, Guangzhou, China
| | - Weijian Bei
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega, Guangzhou, China
| | - Jiao Guo
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Science and Technology Building, 280 Waihuan East Road, Guangzhou Higher Education Mega, Guangzhou, China.
| |
Collapse
|
4
|
Zhou C, Lv M, Wang P, Guo C, Ni Z, Bao H, Tang Y, Cai H, Lu J, Deng W, Yang X, Xia G, Wang H, Wang C, Kong S. Sequential activation of uterine epithelial IGF1R by stromal IGF1 and embryonic IGF2 directs normal uterine preparation for embryo implantation. J Mol Cell Biol 2021; 13:646-661. [PMID: 34097060 PMCID: PMC8648386 DOI: 10.1093/jmcb/mjab034] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/07/2021] [Accepted: 03/17/2021] [Indexed: 11/29/2022] Open
Abstract
Embryo implantation in both humans and rodents is initiated by the attachment of a blastocyst to the uterine epithelium. For blastocyst attachment, the uterine epithelium needs to transform at both the structural and molecular levels first, and then initiate the interaction with trophectoderm. Any perturbation during this process will result in implantation failure or long-term adverse pregnancy outcomes. Endocrine steroid hormones, which function through nuclear receptors, combine with the local molecules produced by the uteri or embryo to facilitate implantation. The insulin-like growth factor (IGF) signaling has been reported to play a vital role during pregnancy. However, its physiological function during implantation remains elusive. This study revealed that mice with conditional deletion of Igf1r gene in uteri suffered from subfertility, mainly due to the disturbed uterine receptivity and abnormal embryo implantation. Mechanistically, we uncovered that in response to the nidatory estrogen on D4 of pregnancy, the epithelial IGF1R, stimulated by the stromal cell-produced IGF1, facilitated epithelial STAT3 activation to modulate the epithelial depolarity. Furthermore, embryonic derived IGF2 could activate both the epithelial ERK1/2 and STAT3 signaling through IGF1R, which was critical for the transcription of Cox2 and normal attachment reaction. In brief, our data revealed that epithelial IGF1R was sequentially activated by the uterine stromal IGF1 and embryonic IGF2 to guarantee normal epithelium differentiation during the implantation process.
Collapse
Affiliation(s)
- Chan Zhou
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, China
| | - Meiying Lv
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, China
- Fuzhou Hospital of Traditional Chinese Medicine Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Peike Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, China
| | - Chuanhui Guo
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, China
| | - Zhangli Ni
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, China
| | - Haili Bao
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, China
| | - Yedong Tang
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, China
| | - Han Cai
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, China
| | - Jinhua Lu
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Wenbo Deng
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Xiaoyu Yang
- Fuzhou Hospital of Traditional Chinese Medicine Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Guoliang Xia
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, College of Life Science, NingXia University, Yinchuan, China
| | - Haibin Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Chao Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Shuangbo Kong
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| |
Collapse
|
5
|
Wu D, Wang C, Li T, Wang H, Hu JH, Peng X. [Study on the effect and mechanism of recombinant human intestinal trefoil factor on intestinal mucosal injury and repair in burned mice]. ZHONGHUA SHAO SHANG ZA ZHI = ZHONGHUA SHAOSHANG ZAZHI = CHINESE JOURNAL OF BURNS 2021; 37:811-820. [PMID: 34645146 PMCID: PMC11917260 DOI: 10.3760/cma.j.cn501120-20210412-00125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Objective: To establish an efficient human intestinal trefoil factor (ITF) recombinant expression and purification strategy and to observe the effect of recombinant human ITF (rhITF) on intestinal mucosal injury and repair in burned rats and to explore the mechanism. Methods: The experimental research method was applied. New yeast expression vector pGAPZαA and yeast X33 were used to express recombinant ITF. The protein was purified by metal chelation affinity chromatography and anion and cation exchange chromatography. The rhITF was identified by non-reductive sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and Western-blotting. The rhITF was mixed with pepsin solution and trypsin solution in a volume ratio of 1∶1, respectively. After mixed with pepsin solution for 0.5, 1.0, 1.5, 2.0 h and trypsin solution for 1.0, 2.0, 4.0 h, the stability of rhITF was analyzed with non-reductive SDS-PAGE. One hundred and five male BALB/c mice aged 6-8 weeks were divided into sham injury group (n=30), burn alone group (n=45), and burn+rhITF group (n=30) according to the random number table. Mice in burn alone group and burn+rhITF group were inflicted with 30% total body surface area full-thickness burn on the back, while mice in sham injury group were simulated with burn. After burn, mice in burn+rhITF group were intragastrically administered with rhITF of 1 mg/kg, while mice in the other two groups were given the same amount of normal saline. At post injury hour 24, 15 mice in burn alone group were collected to prepare burn serum, which was used in the cell experiment. On post injury day (PID) 3, 5, and 7, 10 mice in each group were sacrificed to collect the small intestinal tissue. The pathological changes of the intestinal mucosa were observed by hematoxylin-eosin staining, and the activities of diamine oxidase (DAO) and lactic dehydrogenase (LDH) in the intestinal tissue were determined by spectrophotometry and enzyme linked immunosorbent assay. Three batches of human colorectal adenocarcinoma HT-29 cells were taken and divided into negative control group, 25 μg/mL rhITF group, 50 μg/mL rhITF group (n=3), normal control group, burn serum group, burn serum+rhITF group (n=3), and CK869 inhibitor group, CK666 inhibitor group, solvent control group (n=2), respectively, which were dealt with the corresponding treatment. After 12 h of culture, the migration of cells were observed by Transwell experiment. Another 2 batches of HT-29 cells were taken and each batch of cells were divided into normal control group, burn serum group, and burn serum+rhITF group (n=6). After 24 h of culture, the protein expressions of adenosine monophosphate activated protein kinase (AMPK), phosphorylated AMPK (p-AMPK), Ras related C3 botulinum toxin substrate 1 (Rac1), and actin-related protein 2/3 (Arp 2/3) complex, subunit 1B (ARPC1B) in the cells were detected by Western blotting, and the Rac1 activity of the cells was detected by activated magnetic bead pull-down test. Data were statistically analyzed with analysis of variance for factorial design, one-way analysis of variance, and Student-Newman-Keuls test. Results: Totally 82.35 mg rhITF was gathered from per litre of fermentation broth with protein purity up to 98%, and the rhITF had good antigenicity. The rhITF was stable in pepsin solution and trypsin solution, with 45% rhITF remained after 2.0 h in trypsin solution, and there was 90% rhITF remained after 4.0 h in pepsin solution. At each time point post injury, no hyperemia, or edema was observed in intestinal mucosa of mice in sham injury group, the main pathological manifestations of intestinal mucosa in mice of burn alone group were hyperemia, edema, erosion, and hemorrhage, and the main manifestations of intestinal mucosa of mice in burn+rhITF group were hyperemia and edema on PID 3 and 5, which were alleviated on PID 7. Compared with those of burn alone group, the activities of DAO and LDH in intestinal tissue of mice in sham injury group and burn+rhITF group were significantly increased on PID 3, 5, and 7 (P<0.05 or P<0.01 ). After 12 h of culture, the number of cell migration in 25 μg/mL rhITF group was 58±12, which was obviously more than 16±5 in negative control group (P<0.01) and obviously less than 123±9 in 50 μg/mL rhITF group (P<0.05). After 12 h of culture, the number of cell migration in burn serum group was 60±13, which was significantly less than 143±11 in normal control group and 138±8 in burn serum+rhITF group (P<0.05). After 12 h of culture, the number of cell migration in solvent control group was 155±9, which was significantly more than 33±5 in CK666 inhibitor group and 28±5 in CK869 inhibitor group (P<0.01). After 24 h of culture, the protein expressions of AMPK and Rac1 of cells in burn serum group were close to those of normal control group and burn serum+rhITF group (P˃0.05), the protein expression of p-AMPK of cells in burn serum group was significantly higher than that of normal control group and burn serum+rhITF group, respectively (P<0.05 or P<0.01), and the protein expression of ARPC1B of cells in burn serum group was significantly lower than that of normal control group and burn serum+rhITF group (P<0.05). After 24 h of culture, the Rac1 activity of cells in burn serum group was significantly lower than that in normal control group and burn serum+rhITF group, respectively (P<0.05 or P<0.01). Conclusions: The rhITF obtained in this study has high purity and super stability, which can resist extreme pH and hydrolysis of protease and can relieve intestinal mucosal damage in burned mice. The rhITF can promote the migration of intestinal epithelial cells and accelerate the repair of intestinal mucosa through inhibiting phosphorylation of AMPK to maintain Rac1-Arp2/3 activity.
Collapse
Affiliation(s)
- D Wu
- State Key Laboratory of Trauma, Burns, and Combined Injury, Institute of Burn Research, the First Affiliated Hospital of Army Medical University (the Third Military Medical University), Chongqing 400038, China
| | - C Wang
- State Key Laboratory of Trauma, Burns, and Combined Injury, Institute of Burn Research, the First Affiliated Hospital of Army Medical University (the Third Military Medical University), Chongqing 400038, China
| | - T Li
- State Key Laboratory of Trauma, Burns, and Combined Injury, Institute of Burn Research, the First Affiliated Hospital of Army Medical University (the Third Military Medical University), Chongqing 400038, China
| | - H Wang
- State Key Laboratory of Trauma, Burns, and Combined Injury, Institute of Burn Research, the First Affiliated Hospital of Army Medical University (the Third Military Medical University), Chongqing 400038, China
| | - J H Hu
- State Key Laboratory of Trauma, Burns, and Combined Injury, Institute of Burn Research, the First Affiliated Hospital of Army Medical University (the Third Military Medical University), Chongqing 400038, China
| | - X Peng
- State Key Laboratory of Trauma, Burns, and Combined Injury, Institute of Burn Research, the First Affiliated Hospital of Army Medical University (the Third Military Medical University), Chongqing 400038, China
| |
Collapse
|
6
|
Velmurugan BK, Lin JT, Mahalakshmi B, Lin CC, Chuang YC, Lo YS, Ho HY, Hsieh MJ, Chen MK. Dehydrocrenatidine inhibits head and neck cancer cells invasion and migration by modulating JNK1/2 and ERK1/2 pathway and decreases MMP-2 expression. ENVIRONMENTAL TOXICOLOGY 2021; 36:1848-1856. [PMID: 34076342 DOI: 10.1002/tox.23305] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/14/2021] [Accepted: 05/24/2021] [Indexed: 06/12/2023]
Abstract
Head and neck cancer is associated with poor prognosis because of its highly metastatic nature. For the better management of head and neck cancer patients, it is very important to diagnose the cancer at an early stage, as well as to prevent the rapid spread of cancer either through direct invasion or lymphatic metastasis. In present study, the effect of dehydrocrenatidine, which is a beta-carboline alkaloid found in the medicinal plant Picrasma quassioides, on human head and neck cancer metastasis was investigated. The study results revealed the treatment of FaDu, SCC9, and SCC47 cells with 5, 10, and 20 μM of dehydrocrenatidine significantly decreased the motility, migration, and invasion of head and neck cancer cells. Moreover, the dehydrocrenatidine treatment significantly decreased the expression of MMP-2 and phosphorylation of ERK1/2 and JNK1/2. Additional experiments revealed that the cotreatment of dehydrocrenatidine with either ERK1/2 or JNK1/2 inhibitor caused further reduction in cancer cell motility and migration compared to that in dehydrocrenatidine treatment alone. Moreover, similar trend was observed in case of ERK1/2 and JNK1/2 phosphorylation and MMP-2 expression after the cotreatment. Taken together, the mechanism by which dehydrocrenatidine can decrease the phosphorylation of ERK1/2 and JNK1/2, follow decrease the expression of MMP-2 and inhibits head and neck cancer cells invasion and migration. This present study identifies dehydrocrenatidine as a potent antimetastatic agent that can be used clinically to improve head and neck cancer prognosis.
Collapse
Affiliation(s)
| | - Jen-Tsun Lin
- Division of Hematology and Oncology, Department of Medicine, Changhua Christian Hospital, Changhua, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Post Baccalaureate Medicine, National Chung Hsing University, Taichung, Taiwan
| | - B Mahalakshmi
- Department of Research and Development, Vels Publishers, Tamilnadu, India
| | - Chia-Chieh Lin
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Yi-Ching Chuang
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Yu-Sheng Lo
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Hsin-Yu Ho
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Ming-Ju Hsieh
- Post Baccalaureate Medicine, National Chung Hsing University, Taichung, Taiwan
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Mu-Kuan Chen
- Department of Otorhinolaryngology, Head and Neck Surgery, Changhua Christian Hospital, Changhua, Taiwan
| |
Collapse
|
7
|
Braga Emidio N, Meli R, Tran HNT, Baik H, Morisset-Lopez S, Elliott AG, Blaskovich MAT, Spiller S, Beck-Sickinger AG, Schroeder CI, Muttenthaler M. Chemical Synthesis of TFF3 Reveals Novel Mechanistic Insights and a Gut-Stable Metabolite. J Med Chem 2021; 64:9484-9495. [PMID: 34142550 PMCID: PMC8273887 DOI: 10.1021/acs.jmedchem.1c00767] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Indexed: 01/07/2023]
Abstract
TFF3 regulates essential gastro- and neuroprotective functions, but its molecular mode of action remains poorly understood. Synthetic intractability and lack of reliable bioassays and validated receptors are bottlenecks for mechanistic and structure-activity relationship studies. Here, we report the chemical synthesis of TFF3 and its homodimer via native chemical ligation followed by oxidative folding. Correct folding was confirmed by NMR and circular dichroism, and TFF3 and its homodimer were not cytotoxic or hemolytic. TFF3, its homodimer, and the trefoil domain (TFF310-50) were susceptible to gastrointestinal degradation, revealing a gut-stable metabolite (TFF37-54; t1/2 > 24 h) that retained its trefoil structure and antiapoptotic bioactivity. We tried to validate the putative TFF3 receptors CXCR4 and LINGO2, but neither TFF3 nor its homodimer displayed any activity up to 10 μM. The discovery of a gut-stable bioactive metabolite and reliable synthetic accessibility to TFF3 and its analogues are cornerstones for future molecular probe development and structure-activity relationship studies.
Collapse
Affiliation(s)
- Nayara Braga Emidio
- Institute
for Molecular Bioscience, The University
of Queensland, Brisbane, QLD 4072, Australia
| | - Rajeshwari Meli
- Institute
of Biological Chemistry, Faculty of Chemistry, University of Vienna, Vienna 1090, Austria
| | - Hue N. T. Tran
- Institute
for Molecular Bioscience, The University
of Queensland, Brisbane, QLD 4072, Australia
| | - Hayeon Baik
- Institute
of Biological Chemistry, Faculty of Chemistry, University of Vienna, Vienna 1090, Austria
| | - Séverine Morisset-Lopez
- Centre
de Biophysique Moléculaire, CNRS, Unité Propre de Recherche
4301, Université d’Orléans, Orleans 45071, France
| | - Alysha G. Elliott
- Institute
for Molecular Bioscience, The University
of Queensland, Brisbane, QLD 4072, Australia
| | - Mark A. T. Blaskovich
- Institute
for Molecular Bioscience, The University
of Queensland, Brisbane, QLD 4072, Australia
| | - Sabrina Spiller
- Institute
of Biochemistry, Faculty of Life Sciences, Leipzig University, Leipzig 04103, Germany
| | | | - Christina I. Schroeder
- Institute
for Molecular Bioscience, The University
of Queensland, Brisbane, QLD 4072, Australia
- Center
for Cancer Research, National Cancer Institute,
National Institutes of Health, Frederick, Maryland 21702, United States
| | - Markus Muttenthaler
- Institute
for Molecular Bioscience, The University
of Queensland, Brisbane, QLD 4072, Australia
- Institute
of Biological Chemistry, Faculty of Chemistry, University of Vienna, Vienna 1090, Austria
| |
Collapse
|
8
|
Zullo KM, Douglas B, Maloney NM, Ji Y, Wei Y, Herbine K, Cohen R, Pastore C, Cramer Z, Wang X, Wei W, Somsouk M, Hung LY, Lengner C, Kohanski MH, Cohen NA, Herbert DR. LINGO3 regulates mucosal tissue regeneration and promotes TFF2 dependent recovery from colitis. Scand J Gastroenterol 2021; 56:791-805. [PMID: 33941035 PMCID: PMC8647134 DOI: 10.1080/00365521.2021.1917650] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Aim: Recovery of damaged mucosal surfaces following inflammatory insult requires diverse regenerative mechanisms that remain poorly defined. Previously, we demonstrated that the reparative actions of Trefoil Factor 3 (TFF3) depend upon the enigmatic receptor, leucine rich repeat and immunoglobulin-like domain containing nogo receptor 2 (LINGO2). This study examined the related orphan receptor LINGO3 in the context of intestinal tissue damage to determine whether LINGO family members are generally important for mucosal wound healing and maintenance of the intestinal stem cell (ISC) compartment needed for turnover of mucosal epithelium.Methods and Results: We find that LINGO3 is broadly expressed on human enterocytes and sparsely on discrete cells within the crypt niche, that contains ISCs. Loss of function studies indicate that LINGO3 is involved in recovery of normal intestinal architecture following dextran sodium sulfate (DSS)-induced colitis, and that LINGO3 is needed for therapeutic action of the long acting TFF2 fusion protein (TFF2-Fc), including a number of signaling pathways critical for cell proliferation and wound repair. LINGO3-TFF2 protein-protein interactions were relatively weak however and LINGO3 was only partially responsible for TFF2 induced MAPK signaling suggesting additional un-identified components of a receptor complex. However, deficiency in either TFF2 or LINGO3 abrogated budding/growth of intestinal organoids and reduced expression of the intestinal ISC gene leucine-rich repeat-containing G-protein coupled receptor 5 (LGR5), indicating homologous roles for these proteins in tissue regeneration, possibly via regulation of ISCs in the crypt niche.Conclusion: We propose that LINGO3 serves a previously unappreciated role in promoting mucosal wound healing.
Collapse
Affiliation(s)
- Kelly M. Zullo
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Bonnie Douglas
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Nicole M. Maloney
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Yingbiao Ji
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Yun Wei
- Department of Medicine, Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Karl Herbine
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Rachel Cohen
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Christopher Pastore
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Zvi Cramer
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Xin Wang
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Wenjie Wei
- Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19147
| | - Ma Somsouk
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Li Yin Hung
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104,Department of Medicine, Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Christopher Lengner
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Michael H. Kohanski
- Department of Otorhinolaryngology—Head and Neck Surgery, Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA 19104,The Corporal Michael J. Crescenz VA Medical Center Surgical Service, Philadelphia, PA 19104
| | - Noam A. Cohen
- Department of Otorhinolaryngology—Head and Neck Surgery, Perelman School of Medicine at The University of Pennsylvania, Philadelphia, PA 19104,The Corporal Michael J. Crescenz VA Medical Center Surgical Service, Philadelphia, PA 19104,Monell Chemical Senses Center, Philadelphia, PA 19104
| | - De’Broski R. Herbert
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104,Department of Medicine, Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
9
|
Goswami S, Balasubramanian I, D'Agostino L, Bandyopadhyay S, Patel R, Avasthi S, Yu S, Goldenring JR, Bonder EM, Gao N. RAB11A-mediated YAP localization to adherens and tight junctions is essential for colonic epithelial integrity. J Biol Chem 2021; 297:100848. [PMID: 34058200 PMCID: PMC8254046 DOI: 10.1016/j.jbc.2021.100848] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 05/21/2021] [Accepted: 05/27/2021] [Indexed: 12/16/2022] Open
Abstract
Within the intestinal epithelium, regulation of intracellular protein and vesicular trafficking is of utmost importance for barrier maintenance, immune responses, and tissue polarity. RAB11A is a small GTPase that mediates the anterograde transport of protein cargos to the plasma membrane. Loss of RAB11A-dependent trafficking in mature intestinal epithelial cells results in increased epithelial proliferation and nuclear accumulation of Yes-associated protein (YAP), a key Hippo-signaling transducer that senses cell–cell contacts and regulates tissue growth. However, it is unclear how RAB11A regulates YAP intracellular localizations. In this report, we examined the relationship of RAB11A to epithelial junctional complexes, YAP, and the associated consequences on colonic epithelial tissue repair. We found that RAB11A controls the biochemical associations of YAP with multiple components of adherens and tight junctions, including α-catenin, β-catenin, and Merlin, a tumor suppressor. In the absence of RAB11A and Merlin, we observed enhanced YAP–β-catenin complex formation and nuclear translocation. Upon chemical injury to the intestine, mice deficient in RAB11A were found to have reduced epithelial integrity, decreased YAP localization to adherens and tight junctions, and increased nuclear YAP accumulation in the colon epithelium. Thus, RAB11A-regulated trafficking regulates the Hippo–YAP signaling pathway for rapid reparative response after tissue injury.
Collapse
Affiliation(s)
- Sayantani Goswami
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | | | - Luca D'Agostino
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | | | - Radha Patel
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Shail Avasthi
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Shiyan Yu
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - James R Goldenring
- Department of Surgery, Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Edward M Bonder
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Nan Gao
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA.
| |
Collapse
|
10
|
Goggins BJ, Minahan K, Sherwin S, Soh WS, Pryor J, Bruce J, Liu G, Mathe A, Knight D, Horvat JC, Walker MM, Keely S. Pharmacological HIF-1 stabilization promotes intestinal epithelial healing through regulation of α-integrin expression and function. Am J Physiol Gastrointest Liver Physiol 2021; 320:G420-G438. [PMID: 33470153 DOI: 10.1152/ajpgi.00192.2020] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 12/08/2020] [Accepted: 12/14/2020] [Indexed: 01/31/2023]
Abstract
Intestinal epithelia are critical for maintaining gastrointestinal homeostasis. Epithelial barrier injury, causing inflammation and vascular damage, results in inflammatory hypoxia, and thus, healing occurs in an oxygen-restricted environment. The transcription factor hypoxia-inducible factor (HIF)-1 regulates genes important for cell survival and repair, including the cell adhesion protein β1-integrin. Integrins function as αβ-dimers, and α-integrin-matrix binding is critical for cell migration. We hypothesized that HIF-1 stabilization accelerates epithelial migration through integrin-dependent pathways. We aimed to examine functional and posttranslational activity of α-integrins during HIF-1-mediated intestinal epithelial healing. Wound healing was assessed in T84 monolayers over 24 h with/without prolyl-hydroxylase inhibitor (PHDi) (GB-004), which stabilizes HIF-1. Gene and protein expression were measured by RT-PCR and immunoblot, and α-integrin localization was assessed by immunofluorescence. α-integrin function was assessed by antibody-mediated blockade, and integrin α6 regulation was determined by HIF-1α chromatin immunoprecipitation. Models of mucosal wounding and 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced colitis were used to examine integrin expression and localization in vivo. PHDi treatment accelerated wound closure and migration within 12 h, associated with increased integrin α2 and α6 protein, but not α3. Functional blockade of integrins α2 and α6 inhibited PHDi-mediated accelerated wound closure. HIF-1 bound directly to the integrin α6 promoter. PHDi treatment accelerated mucosal healing, which was associated with increased α6 immunohistochemical staining in wound-associated epithelium and wound-adjacent tissue. PHDi treatment increased α6 protein levels in colonocytes of TNBS mice and induced α6 staining in regenerating crypts and reepithelialized inflammatory lesions. Together, these data demonstrate a role for HIF-1 in regulating both integrin α2 and α6 responses during intestinal epithelial healing.NEW & NOTEWORTHY HIF-1 plays an important role in epithelial restitution, selectively inducing integrins α6 and α2 to promote migration and proliferation, respectively. HIF-stabilizing prolyl-hydroxylase inhibitors accelerate intestinal mucosal healing by inducing epithelial integrin expression.
Collapse
Affiliation(s)
- Bridie J Goggins
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Gastrointestinal Research Group, University of Newcastle, New South Wales, Australia
- Priority Research Centre for Digestive Health and Neurogastroenterology, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales, Australia
| | - Kyra Minahan
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Gastrointestinal Research Group, University of Newcastle, New South Wales, Australia
- Priority Research Centre for Digestive Health and Neurogastroenterology, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales, Australia
| | - Simonne Sherwin
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Gastrointestinal Research Group, University of Newcastle, New South Wales, Australia
- Priority Research Centre for Digestive Health and Neurogastroenterology, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales, Australia
| | - Wai S Soh
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Gastrointestinal Research Group, University of Newcastle, New South Wales, Australia
- Priority Research Centre for Digestive Health and Neurogastroenterology, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales, Australia
| | - Jennifer Pryor
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Gastrointestinal Research Group, University of Newcastle, New South Wales, Australia
- Priority Research Centre for Digestive Health and Neurogastroenterology, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales, Australia
| | - Jessica Bruce
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Gastrointestinal Research Group, University of Newcastle, New South Wales, Australia
- Priority Research Centre for Digestive Health and Neurogastroenterology, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales, Australia
| | - Gang Liu
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Gastrointestinal Research Group, University of Newcastle, New South Wales, Australia
- Priority Research Centre for Digestive Health and Neurogastroenterology, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales, Australia
| | - Andrea Mathe
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Gastrointestinal Research Group, University of Newcastle, New South Wales, Australia
- Priority Research Centre for Digestive Health and Neurogastroenterology, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales, Australia
| | - Darryl Knight
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Jay C Horvat
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Marjorie M Walker
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Priority Research Centre for Digestive Health and Neurogastroenterology, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales, Australia
| | - Simon Keely
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Gastrointestinal Research Group, University of Newcastle, New South Wales, Australia
- Priority Research Centre for Digestive Health and Neurogastroenterology, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales, Australia
| |
Collapse
|
11
|
Zhang ZQ, Hu XS, Lu YC, Zhang JP, Li WY, Zhang WY, Feng W, Ding DF, Xu JG. MEK1/2 Inhibitor (GDC0623) Promotes Osteogenic Differentiation of Primary Osteoblasts Inhibited by IL-1 β through the MEK-Erk1/2 and Jak/Stat3 Pathways. Int J Endocrinol 2021; 2021:5720145. [PMID: 34976051 PMCID: PMC8716208 DOI: 10.1155/2021/5720145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 12/02/2021] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE We evaluated the effects and mechanisms of GDC0623 on osteogenic differentiation of osteoblasts induced by IL-1β. Methodology. Osteoblasts were treated with 20 ng/ml IL-1β and 0.1 µM GDC0623. Cell proliferation levels were evaluated by the cell counting kit 8 (CCK8), EdU assay, and western blotting [proliferating cell nuclear antigen (PCNA) and Cyclin D1]. Osteoblasts were cultured in an osteogenic induction medium for 1-3 weeks after which their differentiations were assessed by alkaline phosphatase (ALP) staining, Alizarin Red staining, calcium concentration, immunocytochemistry staining, real-time quantitative PCR (RT-qPCR), and immunofluorescence staining. The osteogenesis-associated mechanisms were further evaluated by western blotting using appropriate antibodies. RESULTS Relative to the control group, IL-1β induced the rapid proliferation of osteoblasts and suppressed their osteogenic differentiations by upregulating the activities of MEK-Erk1/2 as well as Jak-Stat3 pathways and by elevating MMP13 and MMP9 levels. However, blocking of the MEK-Erk1/2 signaling pathway by GDC0623 treatment reversed these effects. CONCLUSION Inhibition of Jak-Stat3 pathway by C188-9 downregulated the expression levels of MMP9 and MMP13, activated MEK-Erk1/2 pathway, and inhibited osteogenic differentiation.
Collapse
Affiliation(s)
- Zeng-Qiao Zhang
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao-Shen Hu
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ye-Chen Lu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jun-Peng Zhang
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wen-Yao Li
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei-Yang Zhang
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Wei Feng
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dao-Fang Ding
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jian-Guang Xu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
12
|
Huang Y, Wang MM, Yang ZZ, Ren Y, Zhang W, Sun ZR, Nie SN. Pretreatment with intestinal trefoil factor alleviates stress-induced gastric mucosal damage via Akt signaling. World J Gastroenterol 2020; 26:7619-7632. [PMID: 33505140 PMCID: PMC7789054 DOI: 10.3748/wjg.v26.i48.7619] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/19/2020] [Accepted: 12/06/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Stress-related gastric mucosal damage or ulcer remains an unsolved issue for critically ill patients. Stress ulcer prophylaxis has been part of routine intensive care, but uncertainty and controversy still exist. Co-secreted with mucins, intestinal trefoil factor (ITF) is reported to promote restitution and regeneration of intestinal mucosal epithelium, although the mechanism remains unknown.
AIM To elucidate the protective effects of ITF on gastric mucosa and explore the possible mechanisms.
METHODS We used a rat model of gastric mucosal damage induced by water immersion restraint stress and lipopolysaccharide-treated human gastric epithelial cell line to investigate the potential effects of ITF on damaged gastric mucosa both in vivo and in vitro.
RESULTS ITF promoted the proliferation and migration and inhibited necrosis of gastric mucosal epithelia in vitro. It also preserved the integrity of gastric mucosa by upregulating expressions of occludin and zonula occludens-1. In the rat model, pretreatment with ITF ameliorated the gastric mucosal epithelial damage and facilitated mucosal repair. The protective effects of ITF were confirmed to be exerted via activation of Akt signaling, and the specific inhibitor of Akt signaling LY249002 reversed the protective effects.
CONCLUSION ITF might be a promising candidate for prevention and treatment of stress-induced gastric mucosal damage, and further studies should be undertaken to verify its clinical feasibility.
Collapse
Affiliation(s)
- Yun Huang
- Nantong Hospital Affiliated to Nanjing University of Chinese Medicine, Nantong 226001, Jiangsu Province, China
| | - Meng-Meng Wang
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Zhi-Zhou Yang
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Yi Ren
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Wei Zhang
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Zhao-Rui Sun
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Shi-Nan Nie
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, Jiangsu Province, China
- Department of Emergency Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| |
Collapse
|
13
|
Zhang W, Yan H, Deng Y, Lou J, Zhang P, Cui Q, Sun H, Tang H, Sun Y, Yang J, Li D, Sun Y. Expression profile and bioinformatics analysis of circular RNA in intestinal mucosal injury and repair after severe burns. Cell Biol Int 2020; 44:2570-2587. [PMID: 32910511 DOI: 10.1002/cbin.11464] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 08/25/2020] [Accepted: 09/07/2020] [Indexed: 01/22/2023]
Abstract
Circular RNA (circRNA) is a novel noncoding RNA that is mostly found in humans and animals. Although the flux of circRNA research has increased in recent years, its precise function is still unclear. Some studies demonstrate that circRNAs can function as microRNA (miRNA) sponges involved in the regulation of competitive endogenous RNAs networks and play a crucial role in many biological processes. Other studies show that circRNAs play multiple biological roles in gastrointestinal diseases. However, the expression characteristics and function of circRNA in intestinal mucosal injury and repair after severe burn have not been reported. This study aims to screen differentially expressed circRNAs in intestinal mucosal injury and repair after severe burns and understand their underlying mechanisms. To test our hypothesis that circRNA may play a role in promoting repair in intestinal mucosa injury after severe burns, we collected the intestinal tissues of three severely burned mice and three pseudo-scalded mice and evaluated the expression of circRNAs via microarray analysis. Quantitative real-time polymerase chain reaction was also used to validate the circRNA microarray data by selecting six based on different multiples, original values, and p values. The host genes of all differentially expressed circRNAs and the downstream target genes of six selected DEcircRNAs were identified by Gene Ontology and the Kyoto Encyclopedia of Genes and Genomes pathway analysis. Meanwhile, we also created a circRNA-miRNA-mRNA network to predict the role and function of circRNAs in intestinal mucosal injury and repair after severe burns.
Collapse
Affiliation(s)
- Wenwen Zhang
- Department of Burn Surgery, The Affiliated Huaihai Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Burn Surgery, The 71st Group Army Hospital of PLA, Xuzhou, Jiangsu, China
| | - Hao Yan
- Department of Burn Surgery, The Affiliated Huaihai Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Burn Surgery, The 71st Group Army Hospital of PLA, Xuzhou, Jiangsu, China
| | - Yuequ Deng
- Department of Burn Surgery, The Affiliated Huaihai Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Burn Surgery, The 71st Group Army Hospital of PLA, Xuzhou, Jiangsu, China
| | - Jiaqi Lou
- Department of Burn Surgery, The Affiliated Huaihai Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Burn Surgery, The 71st Group Army Hospital of PLA, Xuzhou, Jiangsu, China
| | - Pan Zhang
- Department of Burn Surgery, The Affiliated Huaihai Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Burn Surgery, The 71st Group Army Hospital of PLA, Xuzhou, Jiangsu, China
| | - Qingwei Cui
- Department of Burn Surgery, The Affiliated Huaihai Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Burn Surgery, The 71st Group Army Hospital of PLA, Xuzhou, Jiangsu, China
| | - Han Sun
- Department of Burn Surgery, The Affiliated Huaihai Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Burn Surgery, The 71st Group Army Hospital of PLA, Xuzhou, Jiangsu, China
| | - Hao Tang
- Department of Burn Surgery, The Affiliated Huaihai Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Burn Surgery, The 71st Group Army Hospital of PLA, Xuzhou, Jiangsu, China
| | - Yuan Sun
- Department of Burn Surgery, The Affiliated Huaihai Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Burn Surgery, The 71st Group Army Hospital of PLA, Xuzhou, Jiangsu, China
| | - Juan Yang
- Department of Burn Surgery, The Affiliated Huaihai Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Burn Surgery, The 71st Group Army Hospital of PLA, Xuzhou, Jiangsu, China
| | - Dan Li
- Department of Burn Surgery, The Affiliated Huaihai Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Burn Surgery, The 71st Group Army Hospital of PLA, Xuzhou, Jiangsu, China
| | - Yong Sun
- Department of Burn Surgery, The Affiliated Huaihai Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Burn Surgery, The 71st Group Army Hospital of PLA, Xuzhou, Jiangsu, China
| |
Collapse
|
14
|
Inhibition of TFF3 Enhances Sensitivity-and Overcomes Acquired Resistance-to Doxorubicin in Estrogen Receptor-Positive Mammary Carcinoma. Cancers (Basel) 2019; 11:cancers11101528. [PMID: 31658702 PMCID: PMC6826976 DOI: 10.3390/cancers11101528] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 10/01/2019] [Accepted: 10/05/2019] [Indexed: 02/07/2023] Open
Abstract
Dose-dependent toxicity and acquired resistance are two major challenges limiting the efficacious treatment of mammary carcinoma (MC) with doxorubicin. Herein, we investigated the function of Trefoil Factor 3 (TFF3) in the sensitivity and acquired resistance of estrogen receptor positive (ER+) MC cells to doxorubicin. Doxorubicin treatment of ER+MC cells increased TFF3 expression. The depletion of TFF3 by siRNA or inhibition with a small molecule TFF3 inhibitor (AMPC) synergistically enhanced the efficacy of doxorubicin in ER+MC through the suppression of doxorubicin-induced AKT activation and enhancement of doxorubicin-induced apoptosis. Elevated expression of TFF3 and increased activation of AKT were also observed using a model of acquired doxorubicin resistance in ER+MC cells. AMPC partially re-sensitized the doxorubicin resistant cells to doxorubicin-induced apoptosis. Indeed, doxorubicin resistant ER + MC cells exhibited increased sensitivity to AMPC as a single agent compared to doxorubicin sensitive cells. In vivo, AMPC attenuated growth of doxorubicin sensitive ER+MC xenografts whereas it produced regression of xenografts generated by doxorubicin resistant ER+MC cells. Hence, TFF3 inhibition may improve the efficacy and reduce required doses of doxorubicin in ER+MC. Moreover, inhibition of TFF3 may also be an effective therapeutic strategy to eradicate doxorubicin resistant ER+MC.
Collapse
|
15
|
Belle NM, Ji Y, Herbine K, Wei Y, Park J, Zullo K, Hung LY, Srivatsa S, Young T, Oniskey T, Pastore C, Nieves W, Somsouk M, Herbert DR. TFF3 interacts with LINGO2 to regulate EGFR activation for protection against colitis and gastrointestinal helminths. Nat Commun 2019; 10:4408. [PMID: 31562318 PMCID: PMC6764942 DOI: 10.1038/s41467-019-12315-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 08/26/2019] [Indexed: 12/14/2022] Open
Abstract
Intestinal epithelial cells (IEC) have important functions in nutrient absorption, barrier integrity, regeneration, pathogen-sensing, and mucus secretion. Goblet cells are a specialized cell type of IEC that secrete Trefoil factor 3 (TFF3) to regulate mucus viscosity and wound healing, but whether TFF3-responsiveness requires a receptor is unclear. Here, we show that leucine rich repeat receptor and nogo-interacting protein 2 (LINGO2) is essential for TFF3-mediated functions. LINGO2 immunoprecipitates with TFF3, co-localizes with TFF3 on the cell membrane of IEC, and allows TFF3 to block apoptosis. We further show that TFF3-LINGO2 interactions disrupt EGFR-LINGO2 complexes resulting in enhanced EGFR signaling. Excessive basal EGFR activation in Lingo2 deficient mice increases disease severity during colitis and augments immunity against helminth infection. Conversely, TFF3 deficiency reduces helminth immunity. Thus, TFF3-LINGO2 interactions de-repress inhibitory LINGO2-EGFR complexes, allowing TFF3 to drive wound healing and immunity.
Collapse
Affiliation(s)
- Nicole Maloney Belle
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, 19140, USA
| | - Yingbiao Ji
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, 19140, USA
| | - Karl Herbine
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, 19140, USA
| | - Yun Wei
- Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA, 94110, USA.,Department of Inflammation and Oncology, Amgen Inc., 1120 Veterans Boulevard, South San Francisco, CA, 94080, USA
| | - JoonHyung Park
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, 19140, USA
| | - Kelly Zullo
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, 19140, USA
| | - Li-Yin Hung
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, 19140, USA.,Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA, 94110, USA
| | - Sriram Srivatsa
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, 19140, USA
| | - Tanner Young
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, 19140, USA
| | - Taylor Oniskey
- Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA, 94110, USA
| | - Christopher Pastore
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, 19140, USA
| | - Wildaliz Nieves
- Division of Gastroenterology at ZSFG, University of California, San Francisco, San Francisco, CA, 94110, USA
| | - Ma Somsouk
- Division of Gastroenterology at ZSFG, University of California, San Francisco, San Francisco, CA, 94110, USA
| | - De'Broski R Herbert
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, 19140, USA. .,Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA, 94110, USA.
| |
Collapse
|
16
|
Krüger K, Schmid S, Paulsen F, Ignatius A, Klinger P, Hotfiel T, Swoboda B, Gelse K. Trefoil Factor 3 (TFF3) Is Involved in Cell Migration for Skeletal Repair. Int J Mol Sci 2019; 20:ijms20174277. [PMID: 31480518 PMCID: PMC6747154 DOI: 10.3390/ijms20174277] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 08/28/2019] [Accepted: 08/29/2019] [Indexed: 12/25/2022] Open
Abstract
The aim of the study was to explore the possible role of Trefoil Factor Family peptide 3 (TFF3) for skeletal repair. The expression of TFF3 was analyzed in human joint tissues as well as in a murine bone fracture model. Serum levels of TFF3 following a defined skeletal trauma in humans were determined by ELISA. The mRNA expression of TFF3 was analyzed under normoxia and hypoxia. Expression analysis after stimulation of human mesenchymal progenitor cells (MPCs) with TFF3 was performed by RT2 Profiler PCR Array. The effect of recombinant human (rh)TFF3 on MPCs was analysed by different migration and chemotaxis assays. The effect on cell motility was also visualized by fluorescence staining of F-Actin. TFF3 was absent in human articular cartilage, but strongly expressed in the subchondral bone and periosteum of adult joints. Strong TFF3 immunoreactivity was also detected in murine fracture callus. Serum levels of TFF3 were significantly increased after skeletal trauma in humans. Expression analysis demonstrated that rhTFF3 significantly decreased mRNA of ROCK1. Wound healing assays showed increased cell migration of MPCs by rhTFF3. The F-Actin cytoskeleton was markedly influenced by rhTFF3. Cell proliferation was not increased by rhTFF3. The data demonstrate elevated expression of TFF3 after skeletal trauma. The stimulatory effects on cell motility and migration of MPCs suggest a role of TFF3 in skeletal repair.
Collapse
Affiliation(s)
- Katharina Krüger
- Department of Orthopaedic Trauma Surgery, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Sebastian Schmid
- Department of Orthopaedic Trauma Surgery, University Hospital Erlangen, 91054 Erlangen, Germany
- Institute of Functional and Clinical Anatomy, University of Erlangen-Nuernberg, 91054 Erlangen, Germany
| | - Friedrich Paulsen
- Institute of Functional and Clinical Anatomy, University of Erlangen-Nuernberg, 91054 Erlangen, Germany
| | - Anita Ignatius
- Institute of Orthopaedic Research and Biomechanics, University of Ulm, 89081 Ulm, Germany
| | - Patricia Klinger
- Department of Orthopaedic Trauma Surgery, University Hospital Erlangen, 91054 Erlangen, Germany
- Institute of Functional and Clinical Anatomy, University of Erlangen-Nuernberg, 91054 Erlangen, Germany
| | - Thilo Hotfiel
- Division of Orthopaedic Rheumatology, Department of Orthopaedics, University of Erlangen-Nuernberg, 91054 Erlangen, Germany
| | - Bernd Swoboda
- Division of Orthopaedic Rheumatology, Department of Orthopaedics, University of Erlangen-Nuernberg, 91054 Erlangen, Germany
| | - Kolja Gelse
- Department of Orthopaedic Trauma Surgery, University Hospital Erlangen, 91054 Erlangen, Germany.
| |
Collapse
|
17
|
Gao S, Hu J, Wu X, Liang Z. PMA treated THP-1-derived-IL-6 promotes EMT of SW48 through STAT3/ERK-dependent activation of Wnt/β-catenin signaling pathway. Biomed Pharmacother 2018; 108:618-624. [PMID: 30243096 DOI: 10.1016/j.biopha.2018.09.067] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 09/09/2018] [Accepted: 09/11/2018] [Indexed: 12/23/2022] Open
Abstract
Colon cancer is one of the most common digestive malignant tumors that leads to high mortality worldwide, and metastasis is the primary cause of cancer-related death. It is well accepted that the epithelial-mesenchymal transition (EMT) plays a key role in the process of metastasis. As a cytokine that macrophage secretes, IL-6 is involved in the progression of tumors, including the invasion and metastasis via kinds of signaling pathways. However, the mechanism of interactions between IL-6, macrophage, EMT and colon cancer is not fully understood. Increased CD68+ macrophages and IL-6 level were found in colon tumor as compared to normal colon tissue. Metastatic lymph node showed even more CD68+ macrophages and higher IL-6 level than the primary tumor. These results suggested that macrophages and IL-6 play an important role in EMT of colon cancer. In order to investigate the effect of macrophage and IL-6 on EMT of colon cancer, we cultured human colon carcinoma cell line SW48 with conditioned medium (CM) from PMA-stimulated monocyte THP-1 cells and tested for IL-6 dependent EMT pathways. Wound healing assay and Transwell assay were used to analyze cell migration and invasion. Results showed that CM-treated SW48 cells increased IL-6 production and displayed elevated capacity of migration and invasion compared to untreated cells. Increased expressions of EMT markers (N-cadherin, Vimentin and β-catenin) and decreased expression of EMT marker(E-cadherin) were found in CM-treated SW48 cells by Western Blot. The addition of an anti-IL-6 antibody significantly inhibited the increase of EMT markers (Vimentin and β-catenin) as well as cell migration and invasion, suggesting that IL-6 played a critical role in promoting EMT of CM-treated SW48 cells. In addition, we found that the levels of p-STAT3 and p-ERK increased in CM-treated SW48 compared to untreated cells, which can be reversed by AG490, an inhibitor of JAK. In the meantime, the suppression of JAK-associated signaling pathways caused a decrease of β-catenin. In summary, our study suggested that macrophage-induced IL-6 promotes migration and invasion of colon cancer cell via Wnt/β-catenin pathway in STAT3/ERK-dependent way.
Collapse
Affiliation(s)
- Sikang Gao
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Junwu Hu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiongwen Wu
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhihui Liang
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
18
|
Liu J, Kim SY, Shin S, Jung SH, Yim SH, Lee JY, Lee SH, Chung YJ. Overexpression of TFF3 is involved in prostate carcinogenesis via blocking mitochondria-mediated apoptosis. Exp Mol Med 2018; 50:1-11. [PMID: 30139961 PMCID: PMC6107499 DOI: 10.1038/s12276-018-0137-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 05/10/2018] [Accepted: 06/07/2018] [Indexed: 12/24/2022] Open
Abstract
The overexpression of trefoil factor family 3 (TFF3) is observed in a variety of cancers, including prostate cancer (PCa), and its potential role in carcinogenesis, such as activating the PI3K/AKT pathway, is suggested. However, its role and its related mechanisms in prostate tumorigenesis remain unknown. To elucidate the role of TFF3 overexpression in PCa, we silenced TFF3 in two PCa cell lines that overexpressed TFF3 and explored the molecular mechanism behind its antiapoptotic role. We also examined TFF3 expression in 108 Korean PCa specimens and 106 normal prostate tissues by immunohistochemistry (IHC) analysis. The mean TFF3 IHC score in the tumor tissues was significantly higher than that in the normal tissues (4.702 vs. 0.311, P = 2.52 × 10-24). TFF3-silenced cells showed suppressed tumor cell growth and migration. TFF3 silencing decreased BCL2 and increased BAX expression. The translocation of BAX to the mitochondria was also confirmed. After TFF3 silencing, the expression of the mitochondrial proapoptotic proteins, cytochrome C and Smac/DIABLO, was elevated, and these proteins were released from the mitochondria to the cytosol. Downstream mediators of mitochondrial apoptosis, including cleaved caspase-3, caspase-9, and PARP, were also elevated. Accordingly, the proportion of apoptotic cells was significantly higher among TFF3-silenced cells. There was no difference in extrinsic apoptosis-related molecules after TFF3 silencing. All the results support that TFF3 silencing induces the downstream signaling pathway of mitochondria-mediated apoptosis. This study provides a better understanding of the mechanism of prostate tumorigenesis, suggesting TFF3 as a potential biomarker and therapeutic target of PCa.
Collapse
Affiliation(s)
- Jieying Liu
- Precision Medicine Research Center, Seoul, Korea
- Integrated Research Center for Genome Polymorphism, Seoul, Korea
| | - So Youn Kim
- Precision Medicine Research Center, Seoul, Korea
- Integrated Research Center for Genome Polymorphism, Seoul, Korea
| | - Sun Shin
- Precision Medicine Research Center, Seoul, Korea
- Integrated Research Center for Genome Polymorphism, Seoul, Korea
- Department of Microbiology, The Catholic University of Korea, Seoul, Korea
| | - Seung-Hyun Jung
- Precision Medicine Research Center, Seoul, Korea
- Integrated Research Center for Genome Polymorphism, Seoul, Korea
- Cancer Evolution Research Center, Seoul, Korea
| | - Seon-Hee Yim
- Integrated Research Center for Genome Polymorphism, Seoul, Korea
| | - Ji Youl Lee
- Department of Urology, The Catholic University of Korea, Seoul, Korea
| | - Sug-Hyung Lee
- Cancer Evolution Research Center, Seoul, Korea
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yeun-Jun Chung
- Precision Medicine Research Center, Seoul, Korea.
- Integrated Research Center for Genome Polymorphism, Seoul, Korea.
- Department of Microbiology, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
19
|
Tanaka K, Sugiyama H, Yamanari T, Mise K, Morinaga H, Kitagawa M, Onishi A, Ogawa-Akiyama A, Tanabe K, Eguchi J, Ohmoto Y, Shikata K, Wada J. Renal expression of trefoil factor 3 mRNA in association with tubulointerstitial fibrosis in IgA nephropathy. Nephrology (Carlton) 2018; 23:855-862. [PMID: 29987860 PMCID: PMC6174951 DOI: 10.1111/nep.13444] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2018] [Indexed: 12/13/2022]
Abstract
Aim Trefoil factor 3 (TFF3) is a small peptide that is involved in mucosal protection. TFF3 is widely expressed in multiple tissues including kidney tissue. Previous studies have reported that the levels of urinary TFF3 are significantly increased in patients with chronic kidney disease. The aim of this study is to detect the TFF3 mRNA in kidney and elucidate the relationship between renal TFF3 mRNA and tubulointerstitial fibrosis in IgA nephropathy (IgAN). Methods We investigated the renal mRNA expression of TFF3 by real‐time PCR analysis in biopsy specimens from patients with IgAN, other glomerulonephritis (OGN) and minor glomerular abnormalities (MGA). We also determined the renal localization of TFF3 and the levels of urinary TFF3 by immunostaining and ELISA, respectively. Results The renal TFF3 mRNA expression was significantly associated with the urinary TFF3 secretion and the tubulointerstitial fibrosis score in the IgAN group alone. Immunostaining of the renal specimen of IgAN patients revealed that TFF3 is located in the renal tubular epithelial cells. The locations were almost the same as those that showed uromodulin positivity; specifically, the thick ascending limb (TAL) of the loop of Henle and the early portion of the distal tubule. The urinary TFF3 levels were positively correlated with the levels of urinary biomarkers of tubulointerstitial injury in such patients. Conclusion Renal TFF3 mRNA is associated with renal tubulointerstitial fibrosis in IgAN patients. The TFF3 located in the renal tubular epithelial cells may play a role in the progression of tubulointerstitial fibrosis in IgAN patients. Trefoil factor 3 is a peptide that, generally speaking, has protective functions in epithelial biology. This study reports that TFF3 is increased in IgA nephropathy and correlates with injury. Whether TFF3 is functionally a counter‐regulatory, protective factor or whether its overexpression denotes a pathogenic role remains an outstanding question.
Collapse
Affiliation(s)
- Keiko Tanaka
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hitoshi Sugiyama
- Department of Human Resource Development of Dialysis Therapy for Kidney Disease, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Toshio Yamanari
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Koki Mise
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiroshi Morinaga
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Masashi Kitagawa
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Akifumi Onishi
- Department of Human Resource Development of Dialysis Therapy for Kidney Disease, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Ayu Ogawa-Akiyama
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Katsuyuki Tanabe
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Jun Eguchi
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | | | - Kenichi Shikata
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama, Japan
| | - Jun Wada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
20
|
Chen X, Tong C, Li H, Peng W, Li R, Luo X, Ge H, Ran Y, Li Q, Liu Y, Xiong X, Bai Y, Zhang H, Baker PN, Liu X, Qi H. Dysregulated Expression of RPS4Y1 (Ribosomal Protein S4, Y-Linked 1) Impairs STAT3 (Signal Transducer and Activator of Transcription 3) Signaling to Suppress Trophoblast Cell Migration and Invasion in Preeclampsia. Hypertension 2018; 71:481-490. [PMID: 29378854 DOI: 10.1161/hypertensionaha.117.10250] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 09/13/2017] [Accepted: 12/27/2017] [Indexed: 12/15/2022]
Abstract
Normal placentation and a successful pregnancy depend on appropriate trophoblast cell migration and invasion. Inadequate trophoblast invasion and impaired spiral artery remodeling may lead to pregnancy-related disorders, such as preeclampsia. RPS4Y1 (ribosomal protein S4, Y-linked 1) is a member of the S4E family of ribosomal proteins. In this study, we found that RPS4Y1 levels were upregulated in placental samples collected from preeclamptic patients, when compared with the normotensive pregnant women. In vitro, inhibition of RPS4Y1 induced trophoblast cell invasion, promoted placental explant outgrowth, and increased STAT3 (signal transducer and activator of transcription 3) phosphorylation along with elevated expression of N-cadherin and vimentin. Conversely, overexpression of RPS4Y1 results in reduced trophoblast cell invasion and decreased STAT3 phosphorylation. In addition, the suppression of RPS4Y1 promotes trophoblast cell invasion, which could be abolished by the STAT3 knockdown. Meanwhile, we observed reductions of STAT3 phosphorylation expression in preeclampsia patients. Collectively, these results demonstrate that the level of RPS4Y1 expression may be associated with preeclampsia by affecting trophoblast cell migration and invasion via the STAT3/epithelial-mesenchymal transition pathway.
Collapse
Affiliation(s)
- Xuehai Chen
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.)
| | - Chao Tong
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.)
| | - Haiying Li
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.)
| | - Wei Peng
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.)
| | - Rong Li
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.)
| | - Xin Luo
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.)
| | - Huisheng Ge
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.)
| | - Yuxin Ran
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.)
| | - Qin Li
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.)
| | - Yamin Liu
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.)
| | - Xi Xiong
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.)
| | - Yuxiang Bai
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.)
| | - Hua Zhang
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.)
| | - Philip N Baker
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.)
| | - Xiru Liu
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.).
| | - Hongbo Qi
- From the Department of Obstetrics and Gynecology and Canada-China-New Zealand Joint Laboratory of Maternal and Fetal Medicine, First Affiliated Hospital of Chongqing Medical University, China (X.C., C.T., H.L., W.P., R.L., X.L., H.G., Y.R., Q.L., Y.L., X.X., Y.B., H.Z., X.L., H.Q.); and College of Medicine, Biological Sciences and Psychology, University of Leicester, United Kingdom (P.N.B.)
| |
Collapse
|
21
|
Xu C, Li X, Guo P, Wang J. Hypoxia-Induced Activation of JAK/STAT3 Signaling Pathway Promotes Trophoblast Cell Viability and Angiogenesis in Preeclampsia. Med Sci Monit 2017; 23:4909-4917. [PMID: 29030540 PMCID: PMC5652249 DOI: 10.12659/msm.905418] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background To explore the effect of hypoxic preconditioning on the JAK/STAT3 signaling pathway and its effect on trophoblast cell viability and angiogenesis in preeclampsia (PE). Material/Methods Placental tissues from normal pregnant women and PE patients were collected to detect the expression levels of JAK and STAT3. Trophoblast cells separated from the PE patients were assigned to 4 groups. The expression levels of phosphorylated p-JAK and p-STAT3 were measured by Western blot. Cell viability, colony-forming ability, and cell apoptosis were assessed. The levels of vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), and hepatocyte growth factor (HGF) were determined by enzyme-linked immunosorbent assay (ELISA). Results The expression levels of JAK and STAT3 were higher in the placental tissues of PE patients than in those of normal pregnant women. Compared with the blank group, in the hypoxia group the expression levels of p-JAK and p-STAT3 were increased, cell viability was promoted, the number of colonies was increased, cell apoptosis was inhibited, and the levels of VEGF, bFGF, and HGF were all elevated. However, in comparison with the hypoxia group, the expression levels of p-JAK and p-STAT3 were reduced, the cell viability was inhibited, the colonies were decreased, the levels of VEGF, bFGF, and HGF were all decreased, and cell apoptosis was promoted in the hypoxia + si-JAK group. Conclusions These findings indicate that hypoxic preconditioning may contribute to activation of the JAK/STAT3 signaling pathway, thus promoting trophoblast cell viability and angiogenesis in PE.
Collapse
Affiliation(s)
- Chengfang Xu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China (mainland)
| | - Xuejiao Li
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China (mainland)
| | - Peiling Guo
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China (mainland)
| | - Jia Wang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China (mainland)
| |
Collapse
|
22
|
Yang H, Jiang C, Chen X, He K, Hu Y. Protective effects of sinomenine against LPS-induced inflammation in piglets. Microb Pathog 2017; 110:573-577. [DOI: 10.1016/j.micpath.2017.07.044] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 07/27/2017] [Accepted: 07/27/2017] [Indexed: 12/11/2022]
|
23
|
Patterson AM, Watson AJM. Deciphering the Complex Signaling Systems That Regulate Intestinal Epithelial Cell Death Processes and Shedding. Front Immunol 2017; 8:841. [PMID: 28769935 PMCID: PMC5513916 DOI: 10.3389/fimmu.2017.00841] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 07/04/2017] [Indexed: 01/16/2023] Open
Abstract
Intestinal epithelial cells play a fundamental role in maintaining homeostasis. Shedding of intestinal cells in a controlled manner is critical to maintenance of barrier function. Barrier function is maintained during this shedding process by a redistribution of tight junctional proteins to facilitate closure of the gap left by the shedding cell. However, despite the obvious importance of epithelial cell shedding to gut health, a central question is how the extrusion of epithelial cells is achieved, enabling barrier integrity to be maintained in the healthy gut and restored during inflammation remains largely unanswered. Recent studies have provided evidence that excessive epithelial cell shedding and loss of epithelial barrier integrity is triggered by exposure to lipopolysaccharide or tumor necrosis factor alpha. Subsequent studies have provided evidence of the involvement of specific cellular components and signaling mechanisms as well as the functionality of microbiota that can be either detrimental or beneficial for intestinal barrier integrity. This review will focus on the evidence and decipher how the signaling systems through which the mucosal immune system and microbiota can regulate epithelial cell shedding and how these mechanisms interact to preserve the viability of the epithelium.
Collapse
Affiliation(s)
- Angela M Patterson
- Quadram Institute, Norwich Research Park, Norwich, United Kingdom.,Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Alastair J M Watson
- Quadram Institute, Norwich Research Park, Norwich, United Kingdom.,Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| |
Collapse
|
24
|
Zhao Q, Xu L, Sun X, Zhang K, Shen H, Tian Y, Sun F, Li Y. MFG-E8 overexpression promotes colorectal cancer progression via AKT/MMPs signalling. Tumour Biol 2017; 39:1010428317707881. [PMID: 28653875 DOI: 10.1177/1010428317707881] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Qiujie Zhao
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Lin Xu
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Xiaoyan Sun
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Kai Zhang
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Huimin Shen
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Yanan Tian
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Fengkai Sun
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Yanqing Li
- Department of Gastroenterology, Qilu Hospital, Shandong University, Jinan, China
| |
Collapse
|
25
|
Abstract
Trefoil factor (TFF) peptides, with a 40-amino acid motif and including six conserved cysteine residues that form intramolecular disulfide bonds, are a family of mucin-associated secretory molecules mediating many physiological roles that maintain and restore gastrointestinal (GI) mucosal homeostasis. TFF peptides play important roles in response to GI mucosal injury and inflammation. In response to acute GI mucosal injury, TFF peptides accelerate cell migration to seal the damaged area from luminal contents, whereas chronic inflammation leads to increased TFF expression to prevent further progression of disease. Although much evidence supports the physiological significance of TFF peptides in mucosal defenses, the molecular and cellular mechanisms of TFF peptides in the GI epithelium remain largely unknown. In this review, we summarize the functional roles of TFF1, 2, and 3 and illustrate their action mechanisms, focusing on defense mechanisms in the GI tract.
Collapse
Affiliation(s)
- Eitaro Aihara
- Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, Ohio 45267;
| | - Kristen A Engevik
- Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, Ohio 45267;
| | - Marshall H Montrose
- Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, Ohio 45267;
| |
Collapse
|