1
|
Shi M, Su X, Xiang H, Song Q, Yang S. Advances in the mechanism of urinary proteins in calcium oxalate kidney stone formation. Urolithiasis 2025; 53:27. [PMID: 39932538 DOI: 10.1007/s00240-025-01703-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 01/28/2025] [Indexed: 04/30/2025]
Abstract
Kidney stones are a common urological disease worldwide, imposing a significant burden on healthcare systems. Calcium oxalate stones are the predominant form of urinary calculi, with two main theoretical models explaining their pathogenesis: the fixed particle and free particle models. Regardless of the model, the formation of calcium oxalate kidney stones is inseparably linked to crystal nucleation, growth, aggregation, and adhesion in urine. Growing evidence highlights the significant role of urinary proteins, particularly matrix proteins, in the development of calcium oxalate stones. The review classifies urinary proteins impacting calcium oxalate stone formation into three groups: inhibitors, promoters, and dual-regulators, outlining their contributions to the formation process.
Collapse
Affiliation(s)
- Mingcheng Shi
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, People's Republic of China
| | - Xiaozhe Su
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, People's Republic of China
| | - Heng Xiang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, People's Republic of China
| | - Qianlin Song
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, People's Republic of China.
| | - Sixing Yang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, People's Republic of China.
| |
Collapse
|
2
|
Fajardo C RJ, Clavijo C, Díaz GJ, Cadavid LF. Tissue distribution and expression dynamics of trefoil factor genes in the hydroid Hydractinia symbiolongicarpus. Gene 2024; 929:148824. [PMID: 39103057 DOI: 10.1016/j.gene.2024.148824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/26/2024] [Accepted: 08/02/2024] [Indexed: 08/07/2024]
Abstract
Proteins of the trefoil factor family (TFF) participate in mucosal repair and are formed by single or tandemly repeated trefoil domains. TFFs have been extensively studied in mammals and amphibians, but they have not been functionally characterized in other animals. Here we report the identification of two genes expressed in the hydroid Hydractinia symbiolongicarpus, predicted to encode trefoil domain-containing peptides, one with four trefoil domains in tandem and the other one with a trefoil domain flanked by two ShKT domains. Differential expression analyses by qPCR after an immune challenge and an induced mechanical damage, reveal that the former gene (hysyTFF) had no significant changes in expression after the inductions. However, the latter (hysyTFF-like) was overexpressed after three hours post immune challenge and was downregulated after the first hour post epithelial damage. Immunoblot analyses using specific IgY antibodies revealed that hysyTFF is secreted as a high molecular weight complex. Finally, whole mount immunofluorescence assays showed that hysyTFF was predominantly expressed in the endoderm of stolons and polyps, and sparsely in the ectoderm of both polyps and larvae. Thus, the tissue distribution and expression dynamics of trefoil factor genes in H. symbiolongicarpus suggest that hysyTFF is part of an ancient mechanism of epithelial restitution, and the newly reported hysyTFF-like might act as an immune effector gene, perhaps encoding an antibacterial peptide.
Collapse
Affiliation(s)
- R Johana Fajardo C
- Departamento de Biología, Facultad de Ciencias, Universidad Nacional de Colombia - Sede Bogotá, 111321 Bogotá, Colombia
| | - Carlos Clavijo
- Departamento de Biología, Facultad de Ciencias, Universidad Nacional de Colombia - Sede Bogotá, 111321 Bogotá, Colombia
| | - Gonzalo J Díaz
- Departamento de Salud Animal, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional de Colombia - Sede Bogotá, 111321 Bogotá, Colombia
| | - Luis F Cadavid
- Instituto de Genética, Universidad Nacional de Colombia - Sede Bogotá, 111321 Bogotá, Colombia.
| |
Collapse
|
3
|
Erman A, Dragin Jerman U, Peskar D, Šešelja K, Bazina I, Baus Lončar M. Trefoil Factor Protein 3 (TFF3) as a Guardian of the Urinary Bladder Epithelium. J Histochem Cytochem 2024; 72:693-709. [PMID: 39579021 PMCID: PMC11585002 DOI: 10.1369/00221554241299863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 10/28/2024] [Indexed: 11/25/2024] Open
Abstract
Trefoil factor family (TFF) peptides have been examined primarily in the gastrointestinal tract, where they play an important role in the epithelial regeneration. The therapeutic effects of TFFs, particularly the TFF3 protein, have been well studied in humans and in animal models of gastrointestinal injury, whereas little is known about their occurrence and function in the urinary bladder. In this study, we investigated the presence, location, and function of Tff3 in the urinary bladders of wild-type mice (Tff3WT) and compared them with Tff3 knockout mice (Tff3KO) using molecular and microscopic methods at the light and electron microscopic level. Our results show that Tff3 is expressed in the superficial cells of the urothelium, where it colocalizes with the uroplakin UP1b as one of the fundamental structural components of the apical plasma membrane, which is an important component of the blood-urine permeability barrier. Analysis of the urothelium with experimentally induced injury revealed that injury is more severe in Tff3KO mice and urothelial regeneration is attenuated compared with Tff3WT mice, suggesting that Tff3 plays a fine-tuned role in homeostasis and protection of the urothelium. This study provides the first data on the precise location and function of Tff3 in the bladder epithelium. (J Histochem Cytochem XX. XXX-XXX, XXXX).
Collapse
Affiliation(s)
- Andreja Erman
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Urška Dragin Jerman
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Dominika Peskar
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Kate Šešelja
- Laboratory for Neurodegenerative Research, Department of Molecular Medicine, Ruđer Bošković Institute, Zagreb, Croatia
| | - Iva Bazina
- Laboratory for Neurodegenerative Research, Department of Molecular Medicine, Ruđer Bošković Institute, Zagreb, Croatia
| | - Mirela Baus Lončar
- Laboratory for Neurodegenerative Research, Department of Molecular Medicine, Ruđer Bošković Institute, Zagreb, Croatia
| |
Collapse
|
4
|
Wang H, Li J, Wu G, Lin X, Chen J, Liang J, Zhang J, Luo X, Mao H, Xie J, Li Z, Zhou H, Xu K, Yin J, He Y. Activated sympathetic nerve post stroke downregulates Toll-like receptor 5 and disrupts the gut mucosal barrier. Cell Rep Med 2024; 5:101754. [PMID: 39383869 PMCID: PMC11513850 DOI: 10.1016/j.xcrm.2024.101754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 07/27/2024] [Accepted: 09/06/2024] [Indexed: 10/11/2024]
Abstract
The gut permeability significantly increases after ischemic stroke, partly due to disrupted mucosal barrier, but the mechanism remains elusive. Here, we found that the mucus disruption starts at 2 h post stroke, whereas goblet cell functions remain intact. Meanwhile, the flagellated bacteria Helicobacter thrives and penetrates in the mucus layer. Elimination of the mucosal microbiota or transplantation of Helicobacter in germ-free mice reveals an important role of the mucosal microbiota in mucus disruption. The bacterial invasion is due to downregulated Toll-like receptor 5 (TLR5) and its downstream products flagellin-specific IgA and antimicrobial peptides. Knockdown of intestinal TLR5 increases the abundance of flagellated bacteria and exacerbates mucus injury. Intestinal TLR5 is downregulated by the activation of sympathetic nerve. Serum noradrenaline level is positively associated with flagellin level in patients with stroke and patients' prognosis. These findings reveal a neural pathway in which the sympathetic nerve disrupts the mucosal barrier, providing potential therapeutic targets for stroke injury.
Collapse
Affiliation(s)
- Huidi Wang
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Jie Li
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Guangyan Wu
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Xiaofei Lin
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jiaying Chen
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jingru Liang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jiahui Zhang
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Xiaoxia Luo
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Hongyun Mao
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Jiahui Xie
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Zhuang Li
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China
| | - Hongwei Zhou
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China; Guangdong Provincial Clinical Research Center for Laboratory Medicine, Guangzhou, Guangdong 510033, China; State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Kaiyu Xu
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China.
| | - Jia Yin
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Yan He
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, China; Guangdong Provincial Clinical Research Center for Laboratory Medicine, Guangzhou, Guangdong 510033, China; State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, Guangdong 510515, China; Key Laboratory of Mental Health of the Ministry of Education, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
5
|
Lutz F, Han SY, Büyücek S, Möller K, Viehweger F, Schlichter R, Menz A, Luebke AM, Bawahab AA, Reiswich V, Kluth M, Hube-Magg C, Hinsch A, Weidemann S, Lennartz M, Dum D, Bernreuther C, Lebok P, Sauter G, Marx AH, Simon R, Krech T, Fraune C, Gorbokon N, Burandt E, Minner S, Steurer S, Clauditz TS, Jacobsen F. Expression of Trefoil Factor 1 (TFF1) in Cancer: A Tissue Microarray Study Involving 18,878 Tumors. Diagnostics (Basel) 2024; 14:2157. [PMID: 39410561 PMCID: PMC11475926 DOI: 10.3390/diagnostics14192157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/12/2024] [Accepted: 09/21/2024] [Indexed: 10/20/2024] Open
Abstract
Background/Objectives: Trefoil factor 1 (TFF1) plays a role in the mucus barrier. Methods: To evaluate the prevalence of TFF1 expression in cancer, a tissue microarray containing 18,878 samples from 149 tumor types and 608 samples of 76 normal tissue types was analyzed through immunohistochemistry (IHC). Results: TFF1 staining was detectable in 65 of 149 tumor categories. The highest rates of TFF1 positivity were found in mucinous ovarian carcinomas (76.2%), colorectal adenomas and adenocarcinomas (47.1-75%), breast neoplasms (up to 72.9%), bilio-pancreatic adenocarcinomas (42.1-62.5%), gastro-esophageal adenocarcinomas (40.4-50.0%), neuroendocrine neoplasms (up to 45.5%), cervical adenocarcinomas (39.1%), and urothelial neoplasms (up to 24.3%). High TFF1 expression was related to a low grade of malignancy in non-invasive urothelial carcinomas of the bladder (p = 0.0225), low grade of malignancy (p = 0.0003), estrogen and progesterone receptor expression (p < 0.0001), non-triple negativity (p = 0.0005) in invasive breast cancer of no special type, and right-sided tumor location (p = 0.0021) in colorectal adenocarcinomas. Conclusions: TFF1 IHC has only limited utility for the discrimination of different tumor entities given its expression in many tumor entities. The link between TFF1 expression and parameters of malignancy argues for a relevant biological role of TFF1 in cancer. TFF1 may represent a suitable therapeutic target due to its expression in only a few normal cell types.
Collapse
Affiliation(s)
- Florian Lutz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Soo-Young Han
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Seyma Büyücek
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Katharina Möller
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Florian Viehweger
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Ria Schlichter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Anne Menz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Andreas M. Luebke
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Ahmed Abdulwahab Bawahab
- Department of Basic Medical Sciences, College of Medicine, University of Jeddah, Jeddah 21589, Saudi Arabia;
| | - Viktor Reiswich
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Martina Kluth
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Claudia Hube-Magg
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Andrea Hinsch
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Sören Weidemann
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Maximilian Lennartz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - David Dum
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Christian Bernreuther
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Patrick Lebok
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
- Institute of Pathology, Clinical Center Osnabrueck, 49078 Osnabrueck, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Andreas H. Marx
- Department of Pathology, Academic Hospital Fuerth, 90766 Fuerth, Germany;
| | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Till Krech
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
- Institute of Pathology, Clinical Center Osnabrueck, 49078 Osnabrueck, Germany
| | - Christoph Fraune
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
- Institute of Pathology, Clinical Center Osnabrueck, 49078 Osnabrueck, Germany
| | - Natalia Gorbokon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Eike Burandt
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Sarah Minner
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Stefan Steurer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Till S. Clauditz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| | - Frank Jacobsen
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (F.L.); (S.-Y.H.); (S.B.); (K.M.); (F.V.); (R.S.); (A.M.); (A.M.L.); (V.R.); (M.K.); (C.H.-M.); (A.H.); (S.W.); (M.L.); (D.D.); (C.B.); (P.L.); (G.S.); (T.K.); (C.F.); (N.G.); (E.B.); (S.M.); (S.S.); (T.S.C.); (F.J.)
| |
Collapse
|
6
|
Said NM, El-Shaer NH. Association of serum trefoil factor 3 and leptin levels with obesity: A case-control study. Cytokine 2024; 181:156690. [PMID: 38996578 DOI: 10.1016/j.cyto.2024.156690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/21/2024] [Accepted: 07/02/2024] [Indexed: 07/14/2024]
Abstract
BACKGROUND Obesity has a detrimental impact on individuals, communities, and healthcare systems. Trefoil factor 3 is a secretory protein involved in metabolic processes related to weight regulation. However, its relation with obesity is not fully understood. OBJECTIVE We aimed to assess the serum trefoil factor 3 level and to immunohistochemical detect the leptin in obese patients to evaluate their relation to obesity pathogenesis. METHODS As a case-control study, we enrolled 83 non-obese persons as a control group with a BMI (18.5-24.9) and 83 obese persons as a patient group with a BMI > 30. All the study volunteers are subjected to anthropometric measurements, glucose, and lipid profile analysis by colorimetric methods. Serum trefoil factor 3 level was estimated by ELISA and leptin hormone was detected immunohistochemically in the blood using cell block technique. RESULTS ROC curve analysis for TFF3 showed a good relation with obesity with an AUC of 0.891 and a cut-off value of > 96 ng/ml. There was a significant positive correlation between TFF3 and fasting blood sugar, total cholesterol, and triglycerides. The logistic regression analysis showed that TFF3 is a good risk factor for obesity incidence [p = 0.008; OR = 1.117; (95 % CI): 1.029-1.213]. This was confirmed by multiple linear regression that gave an equation for the possibility of predicting BMI using several factors including TFF3 [BMI = 0.821 + 0.051 × TFF3 + 0.044 × FBS + 0.85 × TC]. The more surprising was the ability of the immunohistochemistry cell block technique to detect leptin antigens associated with an obese person blood not only adipose tissue or serum. CONCLUSION Leptin hormone and TFF3 could be good indicators for obesity incidence. Further research with a larger sample size and in different populations could completely approve our results.
Collapse
Affiliation(s)
- Noha Mohamed Said
- Biochemistry Department, Faculty of Science, Zagazig University, Zagazig 44519, Egypt.
| | - Nahla H El-Shaer
- Zoology Department, Faculty of Science, Zagazig University, Zagazig 44519, Egypt.
| |
Collapse
|
7
|
Ethgen LM, Pastore C, Lin C, Reed DR, Hung LY, Douglas B, Sinker D, Herbert DR, Belle NM. A Trefoil factor 3-Lingo2 axis restrains proliferative expansion of type-1 T helper cells during GI nematode infection. Mucosal Immunol 2024; 17:238-256. [PMID: 38336020 PMCID: PMC11086637 DOI: 10.1016/j.mucimm.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 01/22/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024]
Abstract
Host defense at the mucosal interface requires collaborative interactions between diverse cell lineages. Epithelial cells damaged by microbial invaders release reparative proteins such as the Trefoil factor family (TFF) peptides that functionally restore barrier integrity. However, whether TFF peptides and their receptors also serve instructive roles for immune cell function during infection is incompletely understood. Here, we demonstrate that the intestinal trefoil factor, TFF3, restrains (T cell helper) TH1 cell proliferation and promotes host-protective type 2 immunity against the gastrointestinal parasitic nematode Trichuris muris. Accordingly, T cell-specific deletion of the TFF3 receptor, leucine-rich repeat and immunoglobulin containing nogo receptor 2 (LINGO2), impairs TH2 cell commitment, allows proliferative expansion of interferon (IFN)g+ cluster of differentiation (CD)4+ TH1 cells and blocks normal worm expulsion through an IFNg-dependent mechanism. This study indicates that TFF3, in addition to its known tissue reparative functions, drives anti-helminth immunity by controlling the balance between TH1/TH2 subsets.
Collapse
Affiliation(s)
- Lucas M Ethgen
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Christopher Pastore
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Cailu Lin
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
| | - Danielle R Reed
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
| | - Li-Yin Hung
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Bonnie Douglas
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dominic Sinker
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - De'Broski R Herbert
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| | - Nicole M Belle
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
8
|
Cox RM, Wolf JD, Lieberman NA, Lieber CM, Kang HJ, Sticher ZM, Yoon JJ, Andrews MK, Govindarajan M, Krueger RE, Sobolik EB, Natchus MG, Gewirtz AT, deSwart RL, Kolykhalov AA, Hekmatyar K, Sakamoto K, Greninger AL, Plemper RK. Therapeutic mitigation of measles-like immune amnesia and exacerbated disease after prior respiratory virus infections in ferrets. Nat Commun 2024; 15:1189. [PMID: 38331906 PMCID: PMC10853234 DOI: 10.1038/s41467-024-45418-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 01/23/2024] [Indexed: 02/10/2024] Open
Abstract
Measles cases have surged pre-COVID-19 and the pandemic has aggravated the problem. Most measles-associated morbidity and mortality arises from destruction of pre-existing immune memory by measles virus (MeV), a paramyxovirus of the morbillivirus genus. Therapeutic measles vaccination lacks efficacy, but little is known about preserving immune memory through antivirals and the effect of respiratory disease history on measles severity. We use a canine distemper virus (CDV)-ferret model as surrogate for measles and employ an orally efficacious paramyxovirus polymerase inhibitor to address these questions. A receptor tropism-intact recombinant CDV with low lethality reveals an 8-day advantage of antiviral treatment versus therapeutic vaccination in maintaining immune memory. Infection of female ferrets with influenza A virus (IAV) A/CA/07/2009 (H1N1) or respiratory syncytial virus (RSV) four weeks pre-CDV causes fatal hemorrhagic pneumonia with lung onslaught by commensal bacteria. RNAseq identifies CDV-induced overexpression of trefoil factor (TFF) peptides in the respiratory tract, which is absent in animals pre-infected with IAV. Severe outcomes of consecutive IAV/CDV infections are mitigated by oral antivirals even when initiated late. These findings validate the morbillivirus immune amnesia hypothesis, define measles treatment paradigms, and identify priming of the TFF axis through prior respiratory infections as risk factor for exacerbated morbillivirus disease.
Collapse
Affiliation(s)
- Robert M Cox
- Center for Translational Antiviral Research, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, 30303, USA
| | - Josef D Wolf
- Center for Translational Antiviral Research, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, 30303, USA
| | - Nicole A Lieberman
- Virology Division, Department of Laboratory Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Carolin M Lieber
- Center for Translational Antiviral Research, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, 30303, USA
| | - Hae-Ji Kang
- Center for Translational Antiviral Research, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, 30303, USA
| | - Zachary M Sticher
- Emory Institute for Drug Development, Emory University, Atlanta, GA, 30322, USA
| | - Jeong-Joong Yoon
- Center for Translational Antiviral Research, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, 30303, USA
| | - Meghan K Andrews
- Emory Institute for Drug Development, Emory University, Atlanta, GA, 30322, USA
| | | | - Rebecca E Krueger
- Emory Institute for Drug Development, Emory University, Atlanta, GA, 30322, USA
| | - Elizabeth B Sobolik
- Virology Division, Department of Laboratory Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Michael G Natchus
- Emory Institute for Drug Development, Emory University, Atlanta, GA, 30322, USA
| | - Andrew T Gewirtz
- Center for Translational Antiviral Research, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, 30303, USA
| | - Rik L deSwart
- Department of Viroscience, Erasmus MC, Rotterdam, Netherlands
| | | | - Khan Hekmatyar
- Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA, 30303, USA
| | - Kaori Sakamoto
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA
| | - Alexander L Greninger
- Virology Division, Department of Laboratory Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Richard K Plemper
- Center for Translational Antiviral Research, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, 30303, USA.
| |
Collapse
|
9
|
Fey C, Truschel T, Nehlsen K, Damigos S, Horstmann J, Stradal T, May T, Metzger M, Zdzieblo D. Enhancing pre-clinical research with simplified intestinal cell line models. J Tissue Eng 2024; 15:20417314241228949. [PMID: 38449469 PMCID: PMC10916479 DOI: 10.1177/20417314241228949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 01/12/2024] [Indexed: 03/08/2024] Open
Abstract
Two-dimensional culture remains widely employed to determine the bioavailability of orally delivered drugs. To gain more knowledge about drug uptake mechanisms and risk assessment for the patient after oral drug admission, intestinal in vitro models demonstrating a closer similarity to the in vivo situation are needed. In particular, Caco-2 cell-based Transwell® models show advantages as they are reproducible, cost-efficient, and standardized. However, cellular complexity is impaired and cell function is strongly modified as important transporters in the apical membrane are missing. To overcome these limitations, primary organoid-based human small intestinal tissue models were developed recently but the application of these cultures in pre-clinical research still represents an enormous challenge, as culture setup is complex as well as time- and cost-intensive. To overcome these hurdles, we demonstrate the establishment of primary organoid-derived intestinal cell lines by immortalization. Besides exhibiting cellular diversity of the organoid, these immortalized cell lines enable a standardized and more cost-efficient culture. Further, our cell line-based Transwell®-like models display an organ-specific epithelial barrier integrity, ultrastructural features and representative transport functions. Altogether, our novel model systems are cost-efficient with close similarity to the in vivo situation, therefore favoring their use in bioavailability studies in the context of pre-clinical screenings.
Collapse
Affiliation(s)
- Christina Fey
- Translational Center for Regenerative Therapies (TLZ-RT) Würzburg, Branch of the Fraunhofer Institute for Silicate Research (ISC), Würzburg, Germany
| | | | | | - Spyridon Damigos
- Department of Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, Würzburg, Germany
| | - Julia Horstmann
- Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | | | - Marco Metzger
- Translational Center for Regenerative Therapies (TLZ-RT) Würzburg, Branch of the Fraunhofer Institute for Silicate Research (ISC), Würzburg, Germany
- Department of Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, Würzburg, Germany
| | - Daniela Zdzieblo
- Translational Center for Regenerative Therapies (TLZ-RT) Würzburg, Branch of the Fraunhofer Institute for Silicate Research (ISC), Würzburg, Germany
- Department of Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, Würzburg, Germany
- Project Center for Stem Cell Process Engineering (PZ-SPT), Branch of the Fraunhofer Institute for Silicate Research (ISC), Würzburg, Germany
| |
Collapse
|
10
|
Šešelja K, Bazina I, Vrecl M, Farger J, Schicht M, Paulsen F, Baus Lončar M, Pirman T. Tff3 Deficiency Differentially Affects the Morphology of Male and Female Intestines in a Long-Term High-Fat-Diet-Fed Mouse Model. Int J Mol Sci 2023; 24:16342. [PMID: 38003531 PMCID: PMC10671422 DOI: 10.3390/ijms242216342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 11/05/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Trefoil factor family protein 3 (Tff3) protects the gastrointestinal mucosa and has a complex mode of action in different tissues. Here, we aimed to determine the effect of Tff3 deficiency on intestinal tissues in a long-term high-fat-diet (HFD)-fed model. A novel congenic strain without additional metabolically relevant mutations (Tff3-/-/C57Bl6NCrl strain, male and female) was used. Wild type (Wt) and Tff3-deficient mice of both sexes were fed a HFD for 36 weeks. Long-term feeding of a HFD induces different effects on the intestinal structure of Tff3-deficient male and female mice. For the first time, we found sex-specific differences in duodenal morphology. HFD feeding reduced microvilli height in Tff3-deficient females compared to that in Wt females, suggesting a possible effect on microvillar actin filament dynamics. These changes could not be attributed to genes involved in ER and oxidative stress, apoptosis, or inflammation. Tff3-deficient males exhibited a reduced cecal crypt depth compared to that of Wt males, but this was not the case in females. Microbiome-related short-chain fatty acid content was not affected by Tff3 deficiency in HFD-fed male or female mice. Sex-related differences due to Tff3 deficiency imply the need to consider both sexes in future studies on the role of Tff in intestinal function.
Collapse
Affiliation(s)
- Kate Šešelja
- Department of Molecular Medicine, Ruđer Bošković Institute, Bjenička 54, 10 000 Zagreb, Croatia; (K.Š.); (I.B.)
| | - Iva Bazina
- Department of Molecular Medicine, Ruđer Bošković Institute, Bjenička 54, 10 000 Zagreb, Croatia; (K.Š.); (I.B.)
| | - Milka Vrecl
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000 Ljubljana, Slovenia;
| | - Jessica Farger
- Institute of Functional and Clinical Anatomy, Faculty of Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany; (J.F.); (M.S.); (F.P.)
| | - Martin Schicht
- Institute of Functional and Clinical Anatomy, Faculty of Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany; (J.F.); (M.S.); (F.P.)
| | - Friedrich Paulsen
- Institute of Functional and Clinical Anatomy, Faculty of Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany; (J.F.); (M.S.); (F.P.)
| | - Mirela Baus Lončar
- Department of Molecular Medicine, Ruđer Bošković Institute, Bjenička 54, 10 000 Zagreb, Croatia; (K.Š.); (I.B.)
| | - Tatjana Pirman
- Department of Animal Science, Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000 Ljubljana, Slovenia
| |
Collapse
|
11
|
Shin S, Chung YJ, Moon SW, Choi EJ, Kim MR, Chung YJ, Lee SH. Single-cell profiling identifies distinct hormonal, immunologic, and inflammatory signatures of endometriosis-constituting cells. J Pathol 2023; 261:323-334. [PMID: 37807404 DOI: 10.1002/path.6178] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 07/06/2023] [Accepted: 07/20/2023] [Indexed: 10/10/2023]
Abstract
Endometriosis consists of ectopic endometrial epithelial cells (EEECs) and ectopic endometrial stromal cells (EESCs) mixed with heterogeneous stromal cells. To address how endometriosis-constituting cells are different from normal endometrium and among endometriosis subtypes and how their molecular signatures are related to phenotypic manifestations, we analyzed ovarian endometrial cyst (OEC), superficial peritoneal endometriosis (SPE), and deep infiltrating endometriosis (DIE) from 12 patients using single-cell RNA-sequencing (scRNA-seq). We identified 11 cell clusters, including EEEC, EESC, fibroblasts, inflammatory/immune, endothelial, mesothelial, and Schwann cells. For hormonal signatures, EESCs, but not EEECs, showed high estrogen signatures (estrogen response scores and HOXA downregulation) and low progesterone signatures (DKK1 downregulation) compared to normal endometrium. In EEECs, we found MUC5B+ TFF3low cells enriched in endometriosis. In lymphoid cells, evidence for both immune activation (high cytotoxicity in NK) and exhaustion (high checkpoint genes in NKT and cytotoxic T) was identified in endometriosis. Signatures and subpopulations of macrophages were remarkably different among endometriosis subtypes with increased monocyte-derived macrophages and IL1B expression in DIE. The scRNA-seq predicted NRG1 (macrophage)-ERBB3 (Schwann cell) interaction in endometriosis, expressions of which were validated by immunohistochemistry. Myofibroblast subpopulations differed according to the location (OECs from fibroblasts and SPE/DIEs from mesothelial cells and fibroblasts). Endometriosis endothelial cells displayed proinflammation, angiogenesis, and leaky permeability signatures that were enhanced in DIE. Collectively, our study revealed that (1) many cell types-endometrial, lymphoid, macrophage, fibroblast, and endothelial cells-are altered in endometriosis; (2) endometriosis cells show estrogen responsiveness, immunologic cytotoxicity and exhaustion, and proinflammation signatures that are different in endometriosis subtypes; and (3) novel endometriosis-specific findings of MUC5B+ EEECs, mesothelial cell-derived myofibroblasts, and NRG1-ERBB3 interaction may underlie the pathogenesis of endometriosis. Our results may help extend pathologic insights, dissect aggressive diseases, and discover therapeutic targets in endometriosis. © 2023 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Sun Shin
- Departments of Microbiology, The Catholic University of Korea, Seoul, Republic of Korea
- Departments of Precision Medicine Research Center/IRCGP, The Catholic University of Korea, Seoul, Republic of Korea
- Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Youn-Jee Chung
- Departments of Obstetrics and Gynecology, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seong Won Moon
- Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Departments of Pathology, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eun Ji Choi
- Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Departments of Pathology, The Catholic University of Korea, Seoul, Republic of Korea
| | - Mee-Ran Kim
- Departments of Obstetrics and Gynecology, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yeun-Jun Chung
- Departments of Microbiology, The Catholic University of Korea, Seoul, Republic of Korea
- Departments of Precision Medicine Research Center/IRCGP, The Catholic University of Korea, Seoul, Republic of Korea
- Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Departments of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sug Hyung Lee
- Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Departments of Pathology, The Catholic University of Korea, Seoul, Republic of Korea
- Departments of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
12
|
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
|
13
|
Paranjpe I, Jayaraman P, Su CY, Zhou S, Chen S, Thompson R, Del Valle DM, Kenigsberg E, Zhao S, Jaladanki S, Chaudhary K, Ascolillo S, Vaid A, Gonzalez-Kozlova E, Kauffman J, Kumar A, Paranjpe M, Hagan RO, Kamat S, Gulamali FF, Xie H, Harris J, Patel M, Argueta K, Batchelor C, Nie K, Dellepiane S, Scott L, Levin MA, He JC, Suarez-Farinas M, Coca SG, Chan L, Azeloglu EU, Schadt E, Beckmann N, Gnjatic S, Merad M, Kim-Schulze S, Richards B, Glicksberg BS, Charney AW, Nadkarni GN. Proteomic characterization of acute kidney injury in patients hospitalized with SARS-CoV2 infection. COMMUNICATIONS MEDICINE 2023; 3:81. [PMID: 37308534 PMCID: PMC10258469 DOI: 10.1038/s43856-023-00307-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 05/18/2023] [Indexed: 06/14/2023] Open
Abstract
BACKGROUND Acute kidney injury (AKI) is a known complication of COVID-19 and is associated with an increased risk of in-hospital mortality. Unbiased proteomics using biological specimens can lead to improved risk stratification and discover pathophysiological mechanisms. METHODS Using measurements of ~4000 plasma proteins in two cohorts of patients hospitalized with COVID-19, we discovered and validated markers of COVID-associated AKI (stage 2 or 3) and long-term kidney dysfunction. In the discovery cohort (N = 437), we identified 413 higher plasma abundances of protein targets and 30 lower plasma abundances of protein targets associated with COVID-AKI (adjusted p < 0.05). Of these, 62 proteins were validated in an external cohort (p < 0.05, N = 261). RESULTS We demonstrate that COVID-AKI is associated with increased markers of tubular injury (NGAL) and myocardial injury. Using estimated glomerular filtration (eGFR) measurements taken after discharge, we also find that 25 of the 62 AKI-associated proteins are significantly associated with decreased post-discharge eGFR (adjusted p < 0.05). Proteins most strongly associated with decreased post-discharge eGFR included desmocollin-2, trefoil factor 3, transmembrane emp24 domain-containing protein 10, and cystatin-C indicating tubular dysfunction and injury. CONCLUSIONS Using clinical and proteomic data, our results suggest that while both acute and long-term COVID-associated kidney dysfunction are associated with markers of tubular dysfunction, AKI is driven by a largely multifactorial process involving hemodynamic instability and myocardial damage.
Collapse
Affiliation(s)
- Ishan Paranjpe
- Department of Medicine, Stanford University, Stanford, CA, USA
| | - Pushkala Jayaraman
- The Charles Bronfman Institute for Personalized Medicine (CBIPM), Division of Data Driven and Digital Medicine (D3M), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chen-Yang Su
- Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, Canada
- Department of Computer Science, Quantitative Life Sciences, McGill University, Montreal, QC, Canada
| | - Sirui Zhou
- Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, Canada
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC, Canada
| | - Steven Chen
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ryan Thompson
- The Charles Bronfman Institute for Personalized Medicine (CBIPM), Division of Data Driven and Digital Medicine (D3M), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mount Sinai Clinical Intelligence Center (MSCIC), The Charles Bronfman Institute for Personalized Medicine (CBIPM), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Diane Marie Del Valle
- The Mount Sinai Clinical Intelligence Center (MSCIC), The Charles Bronfman Institute for Personalized Medicine (CBIPM), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ephraim Kenigsberg
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shan Zhao
- Department of Anesthesiology, Perioperative and Pain Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Suraj Jaladanki
- The Mount Sinai Clinical Intelligence Center (MSCIC), The Charles Bronfman Institute for Personalized Medicine (CBIPM), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kumardeep Chaudhary
- Clinical Informatics, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India
| | - Steven Ascolillo
- The Mount Sinai Clinical Intelligence Center (MSCIC), The Charles Bronfman Institute for Personalized Medicine (CBIPM), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Akhil Vaid
- The Mount Sinai Clinical Intelligence Center (MSCIC), The Charles Bronfman Institute for Personalized Medicine (CBIPM), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Edgar Gonzalez-Kozlova
- The Mount Sinai Clinical Intelligence Center (MSCIC), The Charles Bronfman Institute for Personalized Medicine (CBIPM), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Justin Kauffman
- The Mount Sinai Clinical Intelligence Center (MSCIC), The Charles Bronfman Institute for Personalized Medicine (CBIPM), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Arvind Kumar
- The Mount Sinai Clinical Intelligence Center (MSCIC), The Charles Bronfman Institute for Personalized Medicine (CBIPM), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manish Paranjpe
- Division of Health Sciences and Technology, Harvard Medical School, Boston, MA, USA
| | - Ross O Hagan
- The Mount Sinai Clinical Intelligence Center (MSCIC), The Charles Bronfman Institute for Personalized Medicine (CBIPM), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Samir Kamat
- The Mount Sinai Clinical Intelligence Center (MSCIC), The Charles Bronfman Institute for Personalized Medicine (CBIPM), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Faris F Gulamali
- The Mount Sinai Clinical Intelligence Center (MSCIC), The Charles Bronfman Institute for Personalized Medicine (CBIPM), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hui Xie
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joceyln Harris
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manishkumar Patel
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kimberly Argueta
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Craig Batchelor
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kai Nie
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sergio Dellepiane
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Leisha Scott
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew A Levin
- The Mount Sinai Clinical Intelligence Center (MSCIC), The Charles Bronfman Institute for Personalized Medicine (CBIPM), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Anesthesiology, Perioperative and Pain Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John Cijiang He
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mayte Suarez-Farinas
- Department of Biostatistics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Steven G Coca
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lili Chan
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Evren U Azeloglu
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric Schadt
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Noam Beckmann
- The Mount Sinai Clinical Intelligence Center (MSCIC), The Charles Bronfman Institute for Personalized Medicine (CBIPM), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sacha Gnjatic
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miram Merad
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Seunghee Kim-Schulze
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brent Richards
- Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, Canada
- Department of Computer Science, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Department of Twin Research, King's College London, London, GB, UK
| | | | - Alexander W Charney
- Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, Canada
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Charles Bronfman Institute for Personalized Medicine (CBIPM), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Girish N Nadkarni
- The Charles Bronfman Institute for Personalized Medicine (CBIPM), Division of Data Driven and Digital Medicine (D3M), Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC, Canada.
- The Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
14
|
Znalesniak EB, Laskou A, Salm F, Haupenthal K, Harder S, Schlüter H, Hoffmann W. The Forms of the Lectin Tff2 Differ in the Murine Stomach and Pancreas: Indications for Different Molecular Functions. Int J Mol Sci 2023; 24:ijms24087059. [PMID: 37108221 PMCID: PMC10138697 DOI: 10.3390/ijms24087059] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 03/30/2023] [Accepted: 04/03/2023] [Indexed: 04/29/2023] Open
Abstract
The lectin TFF2 belongs to the trefoil factor family (TFF). This polypeptide is typically co-secreted with the mucin MUC6 from gastric mucous neck cells, antral gland cells, and duodenal Brunner glands. Here, TFF2 fulfills a protective function by forming a high-molecular-mass complex with the MUC6, physically stabilizing the mucus barrier. In pigs and mice, and slightly in humans, TFF2 is also synthesized in the pancreas. Here, we investigated the murine stomach, pancreas, and duodenum by fast protein liquid chromatography (FPLC) and proteomics and identified different forms of Tff2. In both the stomach and duodenum, the predominant form is a high-molecular-mass complex with Muc6, whereas, in the pancreas, only low-molecular-mass monomeric Tff2 was detectable. We also investigated the expression of Tff2 and other selected genes in the stomach, pancreas, and the proximal, medial, and distal duodenum (RT-PCR analysis). The absence of the Tff2/Muc6 complex in the pancreas is due to a lack of Muc6. Based on its known motogenic, anti-apoptotic, and anti-inflammatory effects, we propose a protective receptor-mediated function of monomeric Tff2 for the pancreatic ductal epithelium. This view is supported by a report that a loss of Tff2 promotes the formation of pancreatic intraductal mucinous neoplasms.
Collapse
Affiliation(s)
- Eva B Znalesniak
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Aikaterini Laskou
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Franz Salm
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Katharina Haupenthal
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Sönke Harder
- Section Mass Spectrometry and Proteomics, Diagnostic Center, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Hartmut Schlüter
- Section Mass Spectrometry and Proteomics, Diagnostic Center, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Werner Hoffmann
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| |
Collapse
|
15
|
Däullary T, Imdahl F, Dietrich O, Hepp L, Krammer T, Fey C, Neuhaus W, Metzger M, Vogel J, Westermann AJ, Saliba AE, Zdzieblo D. A primary cell-based in vitro model of the human small intestine reveals host olfactomedin 4 induction in response to Salmonella Typhimurium infection. Gut Microbes 2023; 15:2186109. [PMID: 36939013 PMCID: PMC10038062 DOI: 10.1080/19490976.2023.2186109] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/21/2023] Open
Abstract
Infection research largely relies on classical cell culture or mouse models. Despite having delivered invaluable insights into host-pathogen interactions, both have limitations in translating mechanistic principles to human pathologies. Alternatives can be derived from modern Tissue Engineering approaches, allowing the reconstruction of functional tissue models in vitro. Here, we combined a biological extracellular matrix with primary tissue-derived enteroids to establish an in vitro model of the human small intestinal epithelium exhibiting in vivo-like characteristics. Using the foodborne pathogen Salmonella enterica serovar Typhimurium, we demonstrated the applicability of our model to enteric infection research in the human context. Infection assays coupled to spatio-temporal readouts recapitulated the established key steps of epithelial infection by this pathogen in our model. Besides, we detected the upregulation of olfactomedin 4 in infected cells, a hitherto unrecognized aspect of the host response to Salmonella infection. Together, this primary human small intestinal tissue model fills the gap between simplistic cell culture and animal models of infection, and shall prove valuable in uncovering human-specific features of host-pathogen interplay.
Collapse
Affiliation(s)
- Thomas Däullary
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg (UKW), Würzburg, Germany
- Faculty of Biology, Biocenter, Chair of Microbiology, Julius-Maximilians-Universität Würzburg (JMU), Würzburg, Germany
| | - Fabian Imdahl
- Helmholtz-Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
| | - Oliver Dietrich
- Helmholtz-Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
| | - Laura Hepp
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg (UKW), Würzburg, Germany
| | - Tobias Krammer
- Helmholtz-Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
| | - Christina Fey
- Fraunhofer Institute for Silicate Research (ISC),Translational Center Regenerative Therapies (TLC-RT), Würzburg, Germany
| | - Winfried Neuhaus
- Austrian Institute of Technology (AIT), Vienna, Austria
- Department of Medicine, Faculty of Medicine and Dentistry, Danube Private University (DPU), Krems, Austria
| | - Marco Metzger
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg (UKW), Würzburg, Germany
- Fraunhofer Institute for Silicate Research (ISC),Translational Center Regenerative Therapies (TLC-RT), Würzburg, Germany
- Fraunhofer Institute for Silicate Research, Project Center for Stem Cell Process Engineering, Würzburg, Germany
| | - Jörg Vogel
- Helmholtz-Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
- Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany
| | - Alexander J Westermann
- Helmholtz-Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
- Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany
| | - Antoine-Emmanuel Saliba
- Helmholtz-Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
| | - Daniela Zdzieblo
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg (UKW), Würzburg, Germany
- Fraunhofer Institute for Silicate Research (ISC),Translational Center Regenerative Therapies (TLC-RT), Würzburg, Germany
- Fraunhofer Institute for Silicate Research, Project Center for Stem Cell Process Engineering, Würzburg, Germany
| |
Collapse
|
16
|
Weste J, Houben T, Harder S, Schlüter H, Lücke E, Schreiber J, Hoffmann W. Different Molecular Forms of TFF3 in the Human Respiratory Tract: Heterodimerization with IgG Fc Binding Protein (FCGBP) and Proteolytic Cleavage in Bronchial Secretions. Int J Mol Sci 2022; 23:ijms232315359. [PMID: 36499686 PMCID: PMC9737082 DOI: 10.3390/ijms232315359] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/25/2022] [Accepted: 11/25/2022] [Indexed: 12/12/2022] Open
Abstract
The polypeptide TFF3 belongs to the trefoil factor family (TFF) of lectins. TFF3 is typically secreted from mucous epithelia together with mucins. Both intestinal and salivary TFF3 mainly exist as disulfide-linked heterodimers with IgG Fc binding protein (FCGBP). Here, we investigated bronchial tissue specimens, bronchial secretions, and bronchoalveolar lavage (BAL) fluid from patients with a chronic obstructive pulmonary disease (COPD) background by fast protein liquid chromatography and proteomics. For the first time, we identified different molecular forms of TFF3 in the lung. The high-molecular mass form represents TFF3-FCGBP oligomers, whereas the low-molecular mass forms are homodimeric and monomeric TFF3 with possibly anti-apoptotic activities. In addition, disulfide-linked TFF3 heterodimers with an Mr of about 60k and 30k were detected in both bronchial secretions and BAL fluid. In these liquids, TFF3 is partly N-terminally truncated probably by neutrophil elastase cleavage. TFF3-FCGBP is likely involved in the mucosal innate immune defense against microbial infections. We discuss a hypothetical model how TFF3 might control FCGBP oligomerization. Furthermore, we did not find indications for interactions of TFF3-FCGBP with DMBT1gp340 or the mucin MUC5AC, glycoproteins involved in mucosal innate immunity. Surprisingly, bronchial MUC5AC appeared to be degraded when compared with gastric MUC5AC.
Collapse
Affiliation(s)
- Jens Weste
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Till Houben
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Sönke Harder
- Section Mass Spectrometry and Proteomics, Diagnostic Center, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Hartmut Schlüter
- Section Mass Spectrometry and Proteomics, Diagnostic Center, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Eva Lücke
- Department of Pneumology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Jens Schreiber
- Department of Pneumology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Werner Hoffmann
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Correspondence:
| |
Collapse
|
17
|
Lin Z, Wan X, Zhang T, Huo H, Zhang X, Li K, Bei W, Guo J, Yang Y. Trefoil factor 3: New highlights in chronic kidney disease research. Cell Signal 2022; 100:110470. [PMID: 36122885 DOI: 10.1016/j.cellsig.2022.110470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/10/2022] [Accepted: 09/13/2022] [Indexed: 11/28/2022]
Abstract
Trefoil factor 3 (TFF3, also known as intestinal trefoil factor) is a small-molecule peptide containing a typical trefoil structure. TFF3 has several biological effects, such as wound healing, immune regulation, neuroprotection, and cell migration and proliferation promotion. Although TFF3 binding sites were identified in rat kidneys more than a decade ago, the specific effects of this small-molecule peptide on kidneys remain unclear. Until recently, much of the research on TFF3 in the kidney field has focused exclusively on its role as a biomarker. Notably, a large prospective randomized study of patients with 29 common clinical diseases revealed that chronic kidney disease (CKD) was associated with the highest serum TFF3 levels, which were 3-fold higher than in acute gastroenteritis, which had the second-highest levels. Examination of each stage of CKD revealed that urine and serum TFF3 levels significantly increased with the progression of CKD. These results suggest that the role of TFF3 in CKD needs further research. The present review summarizes the renal physiological expression, biological functions, and downstream signaling of TFF3, as well as the upstream events that lead to high expression of TFF3 in CKD.
Collapse
Affiliation(s)
- 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
| | - Xiaofen Wan
- 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
| | - Tao Zhang
- 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
| | - Hongyan Huo
- 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
| | - Xiaoyu Zhang
- 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
| | - Kunping Li
- 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
| | - 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.
| |
Collapse
|
18
|
Shekarriz R, Kochaki N, Eslami-Jouibari M, Omrani-Nava V, Ahmadi M, Alizadeh-Navaei R. TFF1 gene single nucleotide polymorphism (rs3761376) and colorectal cancer risk. Mol Biol Rep 2022; 49:10127-10131. [PMID: 36057754 DOI: 10.1007/s11033-022-07828-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 07/29/2022] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Trefoil Factor 1 (TFF1) is a secretory peptide with gastrointestinal protective functions. Abnormal TFF1 expression is reported in some cancers and functional promoter polymorphism in TFF1 is believed to be associated with risk of gastric cancer. We evaluated rs3761376 in a sample of Iranian patients with colorectal cancer. METHODS Peripheral blood samples were taken from pathology confirmed cases of colorectal cancer and healthy volunteers. Genotyping was carried out using Restriction Fragment Length Polymorphism (RFLP) PCR. Any association with clinicopathologic data was assessed by SPSS version 19. RESULTS A total of 245 participants, including 122 patients with cancer and 123 non-cancer subjects were enrolled. Age, body mass index, and smoking habits were not significantly different between the two groups (P > 0.05). Distribution of TFF1 genotypes was not found to be associated with colorectal cancer. However, distant metastasis was more prevalent in carriers of the mutant allele. CONCLUSION TFF1 rs3761376 was not associated with colorectal cancer but it may be involved in metastasis. Therefore, further investigation is warranted to determine this relationship.
Collapse
Affiliation(s)
- Ramin Shekarriz
- Gastrointestinal cancer research center, Non-communicable diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nafiseh Kochaki
- Gastrointestinal cancer research center, Non-communicable diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Eslami-Jouibari
- Gastrointestinal cancer research center, Non-communicable diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Versa Omrani-Nava
- Gastrointestinal cancer research center, Non-communicable diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohadeseh Ahmadi
- Gastrointestinal cancer research center, Non-communicable diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Reza Alizadeh-Navaei
- Gastrointestinal cancer research center, Non-communicable diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
19
|
Paranjpe I, Jayaraman P, Su CY, Zhou S, Chen S, Thompson R, Del Valle DM, Kenigsberg E, Zhao S, Jaladanki S, Chaudhary K, Ascolillo S, Vaid A, Kumar A, Kozlova E, Paranjpe M, O’Hagan R, Kamat S, Gulamali FF, Kauffman J, Xie H, Harris J, Patel M, Argueta K, Batchelor C, Nie K, Dellepiane S, Scott L, Levin MA, He JC, Suarez-Farinas M, Coca SG, Chan L, Azeloglu EU, Schadt E, Beckmann N, Gnjatic S, Merad M, Kim-Schulze S, Richards B, Glicksberg BS, Charney AW, Nadkarni GN. Proteomic Characterization of Acute Kidney Injury in Patients Hospitalized with SARS-CoV2 Infection. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2021.12.09.21267548. [PMID: 36093350 PMCID: PMC9460972 DOI: 10.1101/2021.12.09.21267548] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Acute kidney injury (AKI) is a known complication of COVID-19 and is associated with an increased risk of in-hospital mortality. Unbiased proteomics using biological specimens can lead to improved risk stratification and discover pathophysiological mechanisms. Using measurements of ∼4000 plasma proteins in two cohorts of patients hospitalized with COVID-19, we discovered and validated markers of COVID-associated AKI (stage 2 or 3) and long-term kidney dysfunction. In the discovery cohort (N= 437), we identified 413 higher plasma abundances of protein targets and 40 lower plasma abundances of protein targets associated with COVID-AKI (adjusted p <0.05). Of these, 62 proteins were validated in an external cohort (p <0.05, N =261). We demonstrate that COVID-AKI is associated with increased markers of tubular injury (NGAL) and myocardial injury. Using estimated glomerular filtration (eGFR) measurements taken after discharge, we also find that 25 of the 62 AKI-associated proteins are significantly associated with decreased post-discharge eGFR (adjusted p <0.05). Proteins most strongly associated with decreased post-discharge eGFR included desmocollin-2, trefoil factor 3, transmembrane emp24 domain-containing protein 10, and cystatin-C indicating tubular dysfunction and injury. Using clinical and proteomic data, our results suggest that while both acute and long-term COVID-associated kidney dysfunction are associated with markers of tubular dysfunction, AKI is driven by a largely multifactorial process involving hemodynamic instability and myocardial damage.
Collapse
Affiliation(s)
- Ishan Paranjpe
- The Mount Sinai Clinical Intelligence Center (MSCIC), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Stanford University, San Francisco, California, United States of America
| | - Pushkala Jayaraman
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Division of Data Driven and Digital Medicine (D3M), Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Chen-Yang Su
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Department of Computer Science, McGill University, Montréal, Québec, Canada
| | - Sirui Zhou
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montréal, Québec, Canada
| | - Steven Chen
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ryan Thompson
- The Mount Sinai Clinical Intelligence Center (MSCIC), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Diane Marie Del Valle
- The Mount Sinai Clinical Intelligence Center (MSCIC), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ephraim Kenigsberg
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shan Zhao
- Department of Anesthesiology, Perioperative and Pain Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Suraj Jaladanki
- The Mount Sinai Clinical Intelligence Center (MSCIC), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kumardeep Chaudhary
- The Mount Sinai Clinical Intelligence Center (MSCIC), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Steven Ascolillo
- The Mount Sinai Clinical Intelligence Center (MSCIC), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Akhil Vaid
- The Mount Sinai Clinical Intelligence Center (MSCIC), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Arvind Kumar
- The Mount Sinai Clinical Intelligence Center (MSCIC), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Edgar Kozlova
- The Mount Sinai Clinical Intelligence Center (MSCIC), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manish Paranjpe
- Division of Health Sciences and Technology, Harvard Medical School, Boston, MA, USA
| | - Ross O’Hagan
- The Mount Sinai Clinical Intelligence Center (MSCIC), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Samir Kamat
- The Mount Sinai Clinical Intelligence Center (MSCIC), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Faris F. Gulamali
- The Mount Sinai Clinical Intelligence Center (MSCIC), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Justin Kauffman
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Medicine, Stanford University, San Francisco, California, United States of America
| | - Hui Xie
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joceyln Harris
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manishkumar Patel
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kimberly Argueta
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Craig Batchelor
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kai Nie
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sergio Dellepiane
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Leisha Scott
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew A Levin
- The Mount Sinai Clinical Intelligence Center (MSCIC), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Anesthesiology, Perioperative and Pain Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - John Cijiang He
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Mayte Suarez-Farinas
- Department of Biostatistics, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Steven G Coca
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Lili Chan
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Evren U Azeloglu
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Eric Schadt
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Noam Beckmann
- The Mount Sinai Clinical Intelligence Center (MSCIC), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sacha Gnjatic
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miram Merad
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Seunghee Kim-Schulze
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brent Richards
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montréal, Québec, Canada
- Department of Human Genetics, McGill University, Montréal, Québec, Canada
- Department of Twin Research, King’s College London, London, United Kingdom
| | - Benjamin S Glicksberg
- The Mount Sinai Clinical Intelligence Center (MSCIC), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexander W Charney
- The Mount Sinai Clinical Intelligence Center (MSCIC), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Girish N Nadkarni
- The Mount Sinai Clinical Intelligence Center (MSCIC), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montréal, Québec, Canada
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Division of Data Driven and Digital Medicine (D3M), Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| |
Collapse
|
20
|
Tff3 Deficiency Protects against Hepatic Fat Accumulation after Prolonged High-Fat Diet. LIFE (BASEL, SWITZERLAND) 2022; 12:life12081288. [PMID: 36013467 PMCID: PMC9409972 DOI: 10.3390/life12081288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/09/2022] [Accepted: 08/11/2022] [Indexed: 11/17/2022]
Abstract
Trefoil factor 3 (Tff3) protein is a small secretory protein expressed on various mucosal surfaces and is involved in proper mucosal function and recovery via various mechanisms, including immune response. However, Tff3 is also found in the bloodstream and in various other tissues, including the liver. Its complete attenuation was observed as the most prominent event in the early phase of diabetes in the polygenic Tally Ho mouse model of diabesity. Since then, its role in metabolic processes has emerged. To elucidate the complex role of Tff3, we used a new Tff3-deficient mouse model without additional metabolically relevant mutations (Tff3-/-/C57BL/6NCrl) and exposed it to a high-fat diet (HFD) for a prolonged period (8 months). The effect was observed in male and female mice compared to wild-type (WT) counter groups (n = 10 animals per group). We monitored the animals’ general metabolic parameters, liver morphology, ultrastructure and molecular genes in relevant lipid and inflammatory pathways. Tff3-deficient male mice had reduced body weight and better glucose utilization after 17 weeks of HFD, but longer HFD exposure (32 weeks) resulted in no such change. We found a strong reduction in lipid accumulation in male Tff3-/-/C57BL/6NCrl mice and a less prominent reduction in female mice. This was associated with downregulated peroxisome proliferator-activated receptor gamma (Pparγ) and upregulated interleukin-6 (Il-6) gene expression, although protein level difference did not reach statistical significance due to higher individual variations. Tff3-/-/C57Bl6N mice of both sex had reduced liver steatosis, without major fatty acid content perturbations. Our research shows that Tff3 protein is clearly involved in complex metabolic pathways. Tff3 deficiency in C57Bl6N genetic background caused reduced lipid accumulation in the liver; further research is needed to elucidate its precise role in metabolism-related events.
Collapse
|
21
|
Wang Q, Jiang Y, Du M, Yang L, Yuan Q. Association of functional genetic variants in TFF1 and nephrolithiasis risk in a Chinese population. BMC Urol 2022; 22:127. [PMID: 35987613 PMCID: PMC9392923 DOI: 10.1186/s12894-022-01081-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 07/25/2022] [Indexed: 11/25/2022] Open
Abstract
Trefoil Factor 1 (TFF1) is considered to be able to inhibit the formation of kidney stone. However, genetic variants in TFF1 and corresponding function in kidney stone development are still not well studied. In this study, the discovery set including 230 cases and 250 controls was used to analyze the association between seven tagSNPs of TFF1 gene and the nephrolithiasis risk. Further evaluation was confirmed by the validation set comprising 307 cases and 461 controls. The consequences of the two-stage case–control study indicated that individuals with the rs3761376 A allele have significantly increased nephrolithiasis risk than those with the GG genotypes [adjusted odds ratio (OR) = 1.35, 95% confidence interval (CI) = 1.05–1.73]. Moreover, we also carried out a stratified analysis and found the increased nephrolithiasis risks at A allele among males, overweight individuals, no hypertensive individuals, nondiabetic individuals, smokers, and drinkers. In the following functional experiments, the notably lower expression of TFF1 was exhibited by the vectors carrying A allele compared with those carrying G allele in both luciferase (P = 0.022) and expression vectors (P = 0.041). In addition to tissue detection, we confirmed a significant inverse association of rs3761376 G > A and TFF1 gene expression (P < 0.001). These results suggest that TFF1 rs3761376 may serve as a potential biomarker to predict the risk of nephrolithiasis.
Collapse
|
22
|
Hasebe K, Yamazaki K, Yamaguchi J, Kokuryo T, Yokoyama Y, Miyata K, Fukaya M, Nagino M, Ebata T. Trefoil factor 1 inhibits the development of esophageal adenocarcinoma from Barrett's epithelium. J Transl Med 2022; 102:885-895. [PMID: 35279702 DOI: 10.1038/s41374-022-00771-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 02/05/2022] [Accepted: 02/14/2022] [Indexed: 12/24/2022] Open
Abstract
Trefoil factor family 1 (TFF1) is one of three members of the trefoil factor family that are abundantly expressed in the gastrointestinal mucosal epithelium. Recent studies have shown that TFF1 acts as a tumor suppressor in gastric, pancreatic and hepatocellular carcinogenesis; however, little is known about its function in esophageal carcinogenesis, especially in esophageal adenocarcinoma (EAC). Barrett's epithelium is the metaplastic columnar epithelium of the esophagus and a known premalignant lesion of EAC. To investigate the role of TFF1 in EAC development, a mouse model of Barrett's epithelium was employed, and human specimens of EAC were assessed by immunohistochemistry (IHC) and methylation-specific PCR. Wild-type (WT) mice underwent gastrojejunostomy on the forestomach, resulting in the development of Barrett's epithelium-like (BE-like) epithelium adjacent to the anastomotic site. BE-like epithelium in these mice expressed TFF1, indicating the association of TFF1 with esophageal adenocarcinoma. TFF1-knockout (TFF1KO) mice underwent the same procedure as well, revealing that a deficiency in TFF1 resulted in the development of adenocarcinoma in the anastomotic site, presumably from BE-like epithelium. IHC of human samples revealed strong TFF1 expression in Barrett's epithelium, which was lost in some EACs, confirming the association between TFF1 and EAC development. Aberrant DNA hypermethylation in TFF1 promoter lesions was detected in TFF1-negative human EAC samples, further confirming not only the role of TFF1 in EAC but also the underlying mechanisms of TFF1 regulation. In addition, IHC revealed the nuclear translocation of β-catenin in human and mouse EAC, suggesting that activation of the Wnt/β-catenin pathway was induced by the loss of TFF1. In conclusion, these results indicate that TFF1 functions as a tumor suppressor to inhibit the development of esophageal carcinogenesis from Barrett's epithelium.
Collapse
Affiliation(s)
- Keiji Hasebe
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kimitoshi Yamazaki
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Junpei Yamaguchi
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Toshio Kokuryo
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yukihiro Yokoyama
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazushi Miyata
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahide Fukaya
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masato Nagino
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomoki Ebata
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
23
|
Lacroix G, Gouyer V, Rocher M, Gottrand F, Desseyn JL. A porous cervical mucus plug leads to preterm birth induced by experimental vaginal infection in mice. iScience 2022; 25:104526. [PMID: 35754724 PMCID: PMC9218384 DOI: 10.1016/j.isci.2022.104526] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 03/14/2022] [Accepted: 05/29/2022] [Indexed: 11/12/2022] Open
Abstract
During gestation, the cervical mucus plug (CMP) acts to seal the cervical canal. Pilot studies in humans have suggested that a porous CMP may increase the risk of uterine infection and preterm birth. We examined the gel-forming content of the mouse vagina and the CMP. We experimentally infected pregnant mice by intravaginal administration of pathogens related to preterm birth in humans. We found that the epithelium in both the vagina and cervical canal of pregnant mice produced the two gel-forming mucins Muc5b and Muc5ac. The CMP was porous in Muc5b-deficient mice for which intravaginal administration of Escherichia coli O 55 led to the activation of an inflammatory response in the uterus and 100% preterm births. The pathogen was found in the mucus plug and uterus. This study shows that Muc5b is essential for the in vivo barrier function and the prevention of uterine infections during gestation. Muc5b and Muc5ac are the main gel-forming mucins of the mouse vagina and cervical canal During pregnancy, a cervical mucus plug (CMP) is formed and seals the cervical canal Muc5b-deficient CMP is highly porous Inflammation following vaginal infection causes preterm birth in Muc5b-deficient mice
Collapse
Affiliation(s)
- Guillaume Lacroix
- University Lille, Inserm, CHU Lille, U1286 - Infinite, 59000 Lille, France
| | - Valérie Gouyer
- University Lille, Inserm, CHU Lille, U1286 - Infinite, 59000 Lille, France
| | - Mylène Rocher
- University Lille, Inserm, CHU Lille, U1286 - Infinite, 59000 Lille, France
| | - Frédéric Gottrand
- University Lille, Inserm, CHU Lille, U1286 - Infinite, 59000 Lille, France
| | - Jean-Luc Desseyn
- University Lille, Inserm, CHU Lille, U1286 - Infinite, 59000 Lille, France
| |
Collapse
|
24
|
Rossi HL, Ortiz-Carpena JF, Tucker D, Vaughan AE, Mangalmurti NS, Cohen NA, Herbert DR. Trefoil Factor Family: A Troika for Lung Repair and Regeneration. Am J Respir Cell Mol Biol 2022; 66:252-259. [PMID: 34784491 PMCID: PMC8937240 DOI: 10.1165/rcmb.2021-0373tr] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 11/15/2021] [Indexed: 11/24/2022] Open
Abstract
Tissue damage in the upper and lower airways caused by mechanical abrasion, noxious chemicals, or pathogenic organisms must be followed by rapid restorative processes; otherwise, persistent immunopathology and disease may ensue. This review will discuss evidence for the important role served by trefoil factor (TFF) family members in healthy and diseased airways of humans and rodents. Collectively, these peptides serve to both maintain and restore homeostasis through their regulation of the mucous layer and their control of cell motility, cell differentiation, and immune function in the upper and lower airways. We will also discuss important differences in which trefoil member tracks with homeostasis and disease between humans and mice, which poses a challenge for research in this area. Moreover, we discuss new evidence supporting newly identified receptor binding partners in the leucine-rich repeat and immunoglobulin-like domain-containing NoGo (LINGO) family in mediating the biological effects of TFF proteins in mouse models of epithelial repair and infection. Recent advances in our knowledge regarding TFF peptides suggest that they may be reasonable therapeutic targets in the treatment of upper and lower airway diseases of diverse etiologies. Further work understanding their role in airway homeostasis, repair, and inflammation will benefit from these newly uncovered receptor-ligand interactions.
Collapse
Affiliation(s)
| | | | | | - Andrew E. Vaughan
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania; and
| | | | - Noam A. Cohen
- Department of Otorhinolaryngology: Head and Neck Surgery, Hospital of the University of Philadelphia, Philadelphia, Pennsylvania
| | | |
Collapse
|
25
|
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
|
26
|
Thangaraj SS, Thiesson HC, Svenningsen P, Stubbe J, Palarasah Y, Bistrup C, Jensen BL, Mortensen LA. Mineralocorticoid receptor blockade with spironolactone has no direct effect on plasma IL-17A and injury markers in urine from kidney transplant patients. Am J Physiol Renal Physiol 2021; 322:F138-F149. [PMID: 34894724 DOI: 10.1152/ajprenal.00104.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Kidney transplantation is associated with increased risk of cardiovascular morbidity. Interleukin-17A (IL-17A) mediates kidney injury. Aldosterone promotes T-helper-17 (Th-17) lymphocyte differentiation and IL-17A production through the mineralocorticoid receptor (MR). In this exploratory, post-hoc substudy, it was hypothesized that 1-year intervention with the MR antagonist spironolactone lowers IL-17A and related cytokines and reduces epithelial injury in kidney transplant recipients. Plasma and urine samples were obtained from kidney transplant recipients from a double-blind randomized clinical trial testing spironolactone (n=39) versus placebo (n=41). Plasma concentrations of cytokines IFN-γ, IL-17A, TNF-α, IL-6, IL-1β, and IL-10 were determined before and after 1-year treatment. Urine calbindin, clusterin, KIM-1, osteoactivin, TFF3, and VEGF/creatinine ratios were analyzed. Blood pressure and plasma aldosterone concentration at inclusion did not relate to plasma cytokines and injury markers. None of the cytokines changed in plasma after spironolactone intervention. Plasma IL-17A increased in the placebo group. Spironolactone induced an increase in plasma K+ (0.4 ± 0.4 mmol/L). This increase did not correlate with plasma IL-17A or urine calbindin and TFF3 changes. Ongoing treatment at inclusion with angiotensin-converting-enzyme inhibitor and/or angiotensin II receptor blockers was not associated with changed levels of IL-17A and injury markers and had no effect on the response to spironolactone. Urinary calbindin and TFF3 decreased in the spironolactone group with no difference in between-group analyses. In conclusion, irrespective of ongoing ANGII inhibition, spironolactone has no effect on plasma IL-17A and related cytokines or urinary injury markers in kidney transplant recipients.
Collapse
Affiliation(s)
- Sai Sindhu Thangaraj
- Department of Cardiovascular and Renal Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Helle Charlotte Thiesson
- Department of Nephrology, Odense University Hospital, Odense C, Denmark.,Department of Clinical Research, Faculty of Health Science, University of Southern Denmark
| | - Per Svenningsen
- Department of Cardiovascular and Renal Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Jane Stubbe
- Department of Cardiovascular and Renal Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Yaseelan Palarasah
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of southern Denmark, Odense C, Denmark
| | - Claus Bistrup
- Department of Nephrology, Odense University Hospital, Odense C, Denmark.,Department of Clinical Research, Faculty of Health Science, University of Southern Denmark
| | - Boye L Jensen
- Department of Cardiovascular and Renal Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | |
Collapse
|
27
|
Sugiyama M, Machida N, Yasunaga A, Terai N, Fukasawa H, Ono HK, Kobayashi R, Nishiyama K, Hashimoto O, Kurusu S, Yoshioka K. Vaginal mucus in mice: developmental and gene expression features of epithelial mucous cells during pregnancy†. Biol Reprod 2021; 105:1272-1282. [PMID: 34416757 DOI: 10.1093/biolre/ioab157] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 07/12/2021] [Accepted: 08/16/2021] [Indexed: 01/18/2023] Open
Abstract
The vagina is the site of copulation and serves as the birth canal. It also provides protection against external pathogens. In mice, due to the absence of cervical glands, the vaginal epithelium is the main producer of vaginal mucus. The development and differentiation of vaginal epithelium-constituting cells and the molecular characteristics of vaginal mucus have not been thoroughly examined. Here, we characterized vaginal mucous cell development and the expression of mucus-related factors in pregnant mice. The vaginal mucous epithelium layer thickened and became multilayered after Day 12 of pregnancy and secreted increasing amounts of mucus until early postpartum. Using histochemistry and transmission electron microscopy, we found supra-basal mucous cells as probable candidates for precursor cells. In vaginal mucous cells, the expression of TFF1, a stabilizer of mucus, was high, and some members of mucins and antimicrobial peptides (MUC5B and DEFB1) were expressed in a stage-dependent manner. In summary, this study presents the partial characterization of vaginal epithelial mucous cell lineage and expression of genes encoding several peptide substances that may affect vaginal tissue homeostasis and mucosal immunity during pregnancy and parturition.
Collapse
Affiliation(s)
- Makoto Sugiyama
- Faculty of Veterinary Medicine, Kitasato University School of Veterinary Medicine, Towada, Japan
| | - Nao Machida
- Faculty of Veterinary Medicine, Kitasato University School of Veterinary Medicine, Towada, Japan
| | - Arata Yasunaga
- Faculty of Veterinary Medicine, Kitasato University School of Veterinary Medicine, Towada, Japan.,Department of Animal Science, Kitasato University School of Veterinary Medicine, Towada, Japan
| | - Nanako Terai
- Faculty of Veterinary Medicine, Kitasato University School of Veterinary Medicine, Towada, Japan
| | - Hanae Fukasawa
- Faculty of Veterinary Medicine, Kitasato University School of Veterinary Medicine, Towada, Japan
| | - Hisaya K Ono
- Faculty of Veterinary Medicine, Kitasato University School of Veterinary Medicine, Towada, Japan
| | - Ryosuke Kobayashi
- Laboratory of Genome Science, Biological Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Keita Nishiyama
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Osamu Hashimoto
- Faculty of Bio-Science, Nagahama Institute of Bio-Science and Technology, Nagahama, Japan
| | - Shiro Kurusu
- Faculty of Veterinary Medicine, Kitasato University School of Veterinary Medicine, Towada, Japan
| | - Kazuki Yoshioka
- Faculty of Veterinary Medicine, Kitasato University School of Veterinary Medicine, Towada, Japan
| |
Collapse
|
28
|
Kim SM, Lee HS, Kim MJ, Park HD, Lee SY. Diagnostic Value of Multiple Serum Biomarkers for Vancomycin-Induced Kidney Injury. J Clin Med 2021; 10:5005. [PMID: 34768522 PMCID: PMC8584616 DOI: 10.3390/jcm10215005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/25/2021] [Accepted: 10/25/2021] [Indexed: 12/14/2022] Open
Abstract
Acute kidney injury (AKI) is a major contributor to in-hospital morbidity and mortality. Vancomycin, one of the most commonly used antibiotics in a clinical setting, is associated with AKI, with its incidence ranging up to 43%. Despite the high demand, few studies have investigated serum biomarkers to detect vancomycin-induced kidney injury (VIKI). Here, we evaluated the diagnostic value of nine candidate serum biomarkers for VIKI. A total of 23,182 cases referred for vancomycin concentration measurement from January 2018 to December 2019 were screened and 28 subjects with confirmed VIKI were enrolled (VIKI group). Age- and sex- matched control group consisted of 21 subjects who underwent vancomycin therapy without developing VIKI (non-VIKI group), and 23 healthy controls (HC group). The serum concentrations of clusterin, retinol binding protein 4 (RBP4), interleukin-18 (IL-18), tumor necrosis factor receptor 1 (TNF-R1), C-X-C motif chemokine ligand 10 (CXCL10), neutrophil gelatinase-associated lipocalin (NGAL), osteopontin, trefoil factor-3 (TFF3), and cystatin C were compared among the three groups, and their correlations with estimated glomerular filtration rate (eGFR) and diagnostic values for VIKI were assessed. All of the biomarkers except clusterin and RBP4 exhibited significant elevation in the VIKI group. Serum TFF3, cystatin C, TNF-R1, and osteopontin demonstrated an excellent diagnostic value for VIKI (TFF3, area under the curve (AUC) 0.932; cystatin C, AUC 0.917; TNF-R1, AUC 0.866; osteopontin, AUC 0.787); and except osteopontin, a strong negative correlation with eGFR (TFF3, r = -0.71; cystatin C, r = -0.70; TNF-R1, r = -0.60). IL-18, CXCL10, and NGAL showed weak correlation with eGFR and moderate diagnostic value for VIKI. This study tested multiple serum biomarkers for VIKI and showed that serum TFF3, cystatin C, TNF-R1, and osteopontin could efficiently discriminate VIKI patients. Further studies are warranted to clarify the diagnostic value of these biomarkers in VIKI.
Collapse
Affiliation(s)
- Sang-Mi Kim
- Samsung Medical Center, Department of Laboratory Medicine and Genetics, School of Medicine, Sungkyunkwan University, Seoul 06351, Korea; (S.-M.K.); (H.-S.L.); (H.-D.P.)
| | - Hyun-Seung Lee
- Samsung Medical Center, Department of Laboratory Medicine and Genetics, School of Medicine, Sungkyunkwan University, Seoul 06351, Korea; (S.-M.K.); (H.-S.L.); (H.-D.P.)
| | - Min-Ji Kim
- Biomedical Statistics Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea;
| | - Hyung-Doo Park
- Samsung Medical Center, Department of Laboratory Medicine and Genetics, School of Medicine, Sungkyunkwan University, Seoul 06351, Korea; (S.-M.K.); (H.-S.L.); (H.-D.P.)
| | - Soo-Youn Lee
- Samsung Medical Center, Department of Laboratory Medicine and Genetics, School of Medicine, Sungkyunkwan University, Seoul 06351, Korea; (S.-M.K.); (H.-S.L.); (H.-D.P.)
- Samsung Medical Center, Department of Clinical Pharmacology & Therapeutics, School of Medicine, Sungkyunkwan University, Seoul 06351, Korea
- Department of Health Science and Technology, Samsung Advanced Institute of Health Science and Technology, Sungkyunkwan University, Seoul 06351, Korea
| |
Collapse
|
29
|
Hasebe K, Yamaguchi J, Kokuryo T, Yokoyama Y, Ochiai Y, Nagino M, Ebata T. Trefoil factor family 2 inhibits cholangiocarcinogenesis by regulating the PTEN pathway in mice. Carcinogenesis 2021; 42:1496-1505. [PMID: 34644378 DOI: 10.1093/carcin/bgab093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 10/02/2021] [Accepted: 10/12/2021] [Indexed: 11/14/2022] Open
Abstract
Trefoil factor family 2 (TFF2) is one of three trefoil factor family proteins and is expressed abundantly in the gastrointestinal epithelium. Recent studies have shown that TFF2 acts as a tumor suppressor in gastric and pancreatic carcinogenesis; however, little is known about its function in cholangiocarcinogenesis. To investigate the function of TFF2 in cholangiocellular carcinoma (CCC), immunohistochemistry of surgically resected human CCC samples was performed. TFF2 expression was upregulated in the early stage and lost in the late stage of cholangiocarcinogenesis, suggesting the association of TFF2 and CCC. A TFF2 expression vector was then transfected into a CCC cell line (HuCCT1) in vitro, revealing that TFF2 functions as a tumor suppressor not only by inhibiting proliferation and invasion but also by promoting the apoptosis of cancer cells. In addition, PTEN signaling activity was downregulated by TFF2, suggesting an association between TFF2 and PTEN. Next, hepatic carcinogenesis model mice (KC; albumin-Cre/Lox-Stop-Lox KRAS G12D) were bred with TFF2-knockout mice to generate a TFF2-deficient mouse model (KC/TFF2 -/-). Although the incidence of hepatocellular carcinoma was not different between KC/TFF2 -/- mice and control mice, biliary intraepithelial neoplasm (BilIN), the precursor of CCC, was frequently found in the biliary epithelium of KC/TFF2 -/- mice. Immunohistochemistry revealed that BilIN samples from these mice did not express PTEN. In addition, two KC/TFF2 -/- mice developed CCC adjacent to BilIN, suggesting that TFF2 functions to inhibit the development of CCC in vivo. These results indicate that TFF2 acts as a tumor suppressor to inhibit the development of CCC by regulating PTEN activity.
Collapse
Affiliation(s)
- Keiji Hasebe
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Junpei Yamaguchi
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toshio Kokuryo
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yukihiro Yokoyama
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yosuke Ochiai
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masato Nagino
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomoki Ebata
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
30
|
Minegishi K, Dobashi Y, Tsubochi H, Hagiwara K, Ishibashi Y, Nomura S, Nakamura R, Ohmoto Y, Endo S. TFF-1 Functions to Suppress Multiple Phenotypes Associated with Lung Cancer Progression. Onco Targets Ther 2021; 14:4761-4777. [PMID: 34531663 PMCID: PMC8439977 DOI: 10.2147/ott.s322697] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 08/24/2021] [Indexed: 12/20/2022] Open
Abstract
Introduction Trefoil Factor (TFF) is a member of a protein family comprised of three isoforms, of which TFF-1 exhibits antithetical functions; promotion or suppression of cell proliferation, survival and invasion, depending on the cancer type. However, the pathobiological function of TFF-1 in lung carcinoma has been still unclear. Methods We examined the expression and secretion of TFF-1 using cultured human lung carcinoma cells by immunoblotting, immunofluorescence, enzyme-linked immunosorbent assay and quantitative real-time PCR analyses. The effects of TFF-1 on various phenotypes were analyzed in two cell lines, including those transfected with cDNA encoding TFF-1. Cell proliferation and death were examined by hemocytometer cell counting and by colorimetric viability/cytotoxicity assay. Cell cycle profile, migration and invasion were also examined by flow cytometry, wound healing assay and Matrigel Transwell assay, respectively. The effect of TFF-1 overexpression was confirmed by additional transfection of TFF-1-specific siRNA. Results Endogenous TFF-1 protein expression and secretion into the media were observed exclusively in adenocarcinoma-derived cell lines. Forced overexpression of TFF-1 drove cell cycle transition, while the proliferation decreased by 19% to 25% due to increased cell death. This cell death was predominantly caused by apoptosis, as assessed by the activation of caspase 3/7. Cell migration was also suppressed by 71% to 82% in TFF-1-transfected cells. The suppressive effect of TFF-1 on proliferation and migration was restored by transfection of TFF-1 siRNA. Moreover, invasion was also suppressed to 77% to 83% in TFF-1-transfected cells. Conclusion These findings reveal that TFF-1 functions as a suppressor of cancer proliferation by induction of apoptosis, cell migration and invasion and thus may provide a synergistic target for potential treatment strategies for human lung carcinoma.
Collapse
Affiliation(s)
- Kentaro Minegishi
- Department of Thoracic Surgery, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Yoh Dobashi
- Department of Medicine, Saitama Medical Center, Jichi Medical University, Saitama, Japan.,Department of Pathology, School of Medicine, International University of Health and Welfare, Tochigi, Japan
| | - Hiroyoshi Tsubochi
- Department of Thoracic Surgery, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Koichi Hagiwara
- Division of Pulmonary Medicine, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Yuko Ishibashi
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Division of Breast Surgery, Hospital of the National Center for the Global Health and Medicine, Tokyo, Japan
| | - Sachiyo Nomura
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ritsuko Nakamura
- Department of Molecular and Cellular Pathology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yasukazu Ohmoto
- Tokushima University Industry-University R&D Startup Leading Institute, Tokushima, Japan
| | - Shunsuke Endo
- Department of Thoracic Surgery, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| |
Collapse
|
31
|
Ghanemi A, Yoshioka M, St-Amand J. Trefoil Factor Family Member 2: From a High-Fat-Induced Gene to a Potential Obesity Therapy Target. Metabolites 2021; 11:metabo11080536. [PMID: 34436477 PMCID: PMC8401738 DOI: 10.3390/metabo11080536] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/09/2021] [Accepted: 08/10/2021] [Indexed: 12/11/2022] Open
Abstract
Obesity has its epidemiological patterns continuously increasing. With controlling both diet and exercise being the main approaches to manage the energy metabolism balance, a high-fat (HF) diet is of particular importance. Indeed, lipids have a low satiety potential but a high caloric density. Thus, focusing on pharmacologically targetable pathways remains an approach with promising therapeutic potential. Within this context, trefoil factor family member 2 (Tff2) has been characterized as specifically induced by HF diet rather than low-fat diet. TFF2 has also been linked to diverse neurological mechanisms and metabolic patterns suggesting its role in energy balance. The hypothesis is that TFF2 would be a HF diet-induced signal that regulates metabolism with a focus on lipids. Within this review, we put the spotlight on key findings highlighting this line of thought. Importantly, the hypothetical mechanisms pointed highlight TFF2 as an important contributor to obesity development via increasing lipids intestinal absorption and anabolism. Therefore, an outlook for future experimental activities and evaluation of the therapeutic potential of TFF2 inhibition is given. Indeed, its knockdown or downregulation would contribute to an antiobesity phenotype. We believe this work represents an addition to our understanding of the lipidic molecular implications in obesity, which will contribute to develop therapies aiming to manage the lipidic metabolic pathways including the absorption, storage and metabolism via targeting TFF2-related pathways. We briefly discuss important relevant concepts for both basic and clinical researchers.
Collapse
Affiliation(s)
- Abdelaziz Ghanemi
- Functional Genomics Laboratory, CREMI, Québec Genome Center, CHUL-CHU de Québec Research Center, Quebec, QC G1V 4G2, Canada; (A.G.); (M.Y.)
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec, QC G1V 0A6, Canada
| | - Mayumi Yoshioka
- Functional Genomics Laboratory, CREMI, Québec Genome Center, CHUL-CHU de Québec Research Center, Quebec, QC G1V 4G2, Canada; (A.G.); (M.Y.)
| | - Jonny St-Amand
- Functional Genomics Laboratory, CREMI, Québec Genome Center, CHUL-CHU de Québec Research Center, Quebec, QC G1V 4G2, Canada; (A.G.); (M.Y.)
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec, QC G1V 0A6, Canada
- Correspondence:
| |
Collapse
|
32
|
Samson MH, Abildgaard AM, Espelund U, Rasmussen TR, Folkersen B, Frystyk J, Nexo E. Circulating trefoil factors in relation to lung cancer, age and lung function: a cross-sectional study in patients referred for suspected lung cancer. Scandinavian Journal of Clinical and Laboratory Investigation 2021; 81:446-450. [PMID: 34242119 DOI: 10.1080/00365513.2021.1943757] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The trefoil factor family proteins: TFF1, TFF2 and TFF3 are secreted by epithelial cells in the respiratory tract. Here, we explore circulating concentrations of the trefoil factors in relation to lung cancer, age and lung function. We included 751 patients suspected of lung cancer. Lung cancer diagnosis was based on data reported to a national database. Serum TFF1, TFF2 and TFF3 concentrations were measured by ELISA, and spirometry was performed within ±3 days of blood sampling. Forced expiratory volume in the first second (FEV1) in relation to forced vital capacity (FVC), FEV1/FVC (a parameter used to quantify reduced lung function) was recorded. Lung cancer was diagnosed in 163 (22%) patients. Circulating concentrations of TFF3 (p = .021), but not TFF1 and TFF2, were significantly elevated in cancer patients. All three trefoil factors showed an increase in concentration with increasing age (p < .001) and declining lung function (p < .004). In the present cohort, concentrations of all three peptides were elevated compared with previous results published for healthy individuals. In conclusion, we report higher concentrations of TFF3 in patients with lung cancer, while increasing age and reduced lung function are associated with increasing concentrations of all trefoil factors in this specific patient population. The results emphasize that age and lung function should be taken into consideration when evaluating concentrations of trefoil factors in patients. However, the increases in trefoil factor concentrations were relatively small, and consequently, it is unlikely that circulating trefoil factor concentrations may have a role in the diagnosis of lung cancer and lung function impairment.
Collapse
Affiliation(s)
- Mie H Samson
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Anders M Abildgaard
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Ulrick Espelund
- Department of Clinical Medicine, Medical Research Laboratory, Aarhus University, Aarhus, Denmark
| | - Torben R Rasmussen
- Department of Pulmonary Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Birgitte Folkersen
- Department of Pulmonary Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Jan Frystyk
- Department of Clinical Medicine, Medical Research Laboratory, Aarhus University, Aarhus, Denmark
| | - Ebba Nexo
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
33
|
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
|
34
|
Humulene Inhibits Acute Gastric Mucosal Injury by Enhancing Mucosal Integrity. Antioxidants (Basel) 2021; 10:antiox10050761. [PMID: 34064830 PMCID: PMC8150829 DOI: 10.3390/antiox10050761] [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: 03/16/2021] [Revised: 04/30/2021] [Accepted: 05/07/2021] [Indexed: 12/17/2022] Open
Abstract
This study was designed to determine whether α-humulene, a major constituent in many plants used in fragrances, has a protective role against gastric injury in vivo and in vitro. A rat model of hydrochloric acid (HCl)/ethanol-induced gastritis and human mast cells (HMC-1) were used to investigate the mucosal protective effect of α-humulene. α-Humulene significantly inhibited gastric lesions in HCl/ethanol-induced acute gastritis and decreased gastric acid secretion pyloric ligation-induced gastric ulcers in vivo. In addition, α-humulene reduced the amount of reactive oxygen species and malondialdehyde through upregulation of prostaglandin E2 (PGE2) and superoxide dismutase (SOD). In HMC-1 cells, α-humulene decreased intracellular calcium and increased intracellular cyclic adenosine monophosphate (cAMP) levels, resulting in low histamine levels. α-Humulene also reduced the expression levels of cytokine genes such as interleukin (IL)-1β, IL-6, tumor necrosis factor (TNF) by downregulating nuclear factor-κB (NF-κB) nuclear translocation. Finally, α-humulene upregulated the expression levels of mucin 5AC (Muc5ac), Muc6, trefoil factor 1 (Tff1), trefoil factor 2 (Tff2), and polymeric immunoglobulin receptor (pigr). α-Humulene may attenuate HCl/ethanol-induced gastritis by inhibiting histamine release and NF-κB activation and stimulating antioxidants and mucosal protective factors, particularly Muc5ac and Muc6. Therefore, these data suggest that α-humulene is a potential drug candidate for the treatment of stress-induced or alcoholic gastritis.
Collapse
|
35
|
Hoffmann W. Trefoil Factor Family (TFF) Peptides and Their Links to Inflammation: A Re-evaluation and New Medical Perspectives. Int J Mol Sci 2021; 22:ijms22094909. [PMID: 34066339 PMCID: PMC8125380 DOI: 10.3390/ijms22094909] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/21/2021] [Accepted: 04/21/2021] [Indexed: 12/16/2022] Open
Abstract
Trefoil factor family peptides (TFF1, TFF2, TFF3), together with mucins, are typical exocrine products of mucous epithelia. Here, they act as a gastric tumor suppressor (TFF1) or they play different roles in mucosal innate immune defense (TFF2, TFF3). Minute amounts are also secreted as endocrine, e.g., by the immune and central nervous systems. As a hallmark, TFF peptides have different lectin activities, best characterized for TFF2, but also TFF1. Pathologically, ectopic expression occurs during inflammation and in various tumors. In this review, the role of TFF peptides during inflammation is discussed on two levels. On the one hand, the expression of TFF1-3 is regulated by inflammatory signals in different ways (upstream links). On the other hand, TFF peptides influence inflammatory processes (downstream links). The latter are recognized best in various Tff-deficient mice, which have completely different phenotypes. In particular, TFF2 is secreted by myeloid cells (e.g., macrophages) and lymphocytes (e.g., memory T cells), where it modulates immune reactions triggering inflammation. As a new concept, in addition to lectin-triggered activation, a hypothetical lectin-triggered inhibition of glycosylated transmembrane receptors by TFF peptides is discussed. Thus, TFFs are promising players in the field of glycoimmunology, such as galectins and C-type lectins.
Collapse
Affiliation(s)
- Werner Hoffmann
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| |
Collapse
|
36
|
Ghanemi A, Yoshioka M, St-Amand J. High-Fat Diet-Induced Trefoil Factor Family Member 2 (TFF2) to Counteract the Immune-Mediated Damage in Mice. Animals (Basel) 2021; 11:ani11020258. [PMID: 33494143 PMCID: PMC7909836 DOI: 10.3390/ani11020258] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/16/2021] [Accepted: 01/19/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary High-fat (HF) diet induces both immune-mediated damage and trefoil factor family member 2 (Tff2) expression. As TFF2 has tissue repair and protection properties, this suggests that HF diet-induced Tff2 production and the resulting TFF2 mucosal protective effects would be a mechanism to counteract the HF diet-induced tissue damage. On the other hand, the induction of Tff2 by HF diet could indicate that TFF2 is a food intake regulator (appetite control) since Tff2 is also expressed in the brain. This highlights the importance of exploring TFF2-related pathways in the context of obesity management towards potential therapies. Abstract Physiological homeostasis requires a balance between the immunological functions and the resulting damage/side effects of the immunological reactions including those related to high-fat (HF) diet. Within this context, whereas HF diet, through diverse mechanisms (such as inflammation), leads to immune-mediated damage, trefoil factor family member 2 (Tff2) represents a HF diet-induced gene. On the other hand, TFF2 both promotes tissue repair and reduces inflammation. These properties are towards counteracting the immune-mediated damage resulting from the HF diet. These observations suggest that the HF diet-induction of Tff2 could be a regulatory pathway aiming to counteract the immune-mediated damage resulting from the HF diet. Interestingly, since Tff2 expression increases with HF diet and with Tff2 also expressed in the brain, we also hypothesize that TFF2 could be a HF diet-induced food intake-control signal that reduces appetite. This hypothesis fits with counteracting the immune damage since reducing the food intake will reduce the HF intake and therefore, reduces the HF diet-induced tissue damage. Such food intake signaling would be an indirect mechanism by which TFF2 promotes tissue repair as well as a pathway worth exploring for potential obesity management pharmacotherapies.
Collapse
Affiliation(s)
- Abdelaziz Ghanemi
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec, QC G1V 0A6, Canada;
- Functional Genomics Laboratory, Endocrinology and Nephrology Axis, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada;
| | - Mayumi Yoshioka
- Functional Genomics Laboratory, Endocrinology and Nephrology Axis, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada;
| | - Jonny St-Amand
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec, QC G1V 0A6, Canada;
- Functional Genomics Laboratory, Endocrinology and Nephrology Axis, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada;
- Correspondence: ; Tel.: +1-(418)-525-4444 (ext. 46448); Fax: +1-(418)-654-2298
| |
Collapse
|
37
|
Dodla P, Bhoopalan V, Khoo SK, Miranti C, Sridhar S. Gene expression analysis of human prostate cell lines with and without tumor metastasis suppressor CD82. BMC Cancer 2020; 20:1211. [PMID: 33298014 PMCID: PMC7724878 DOI: 10.1186/s12885-020-07675-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 11/22/2020] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Tetraspanin CD82 is a tumor metastasis suppressor that is known to down regulate in various metastatic cancers. However, the exact mechanism by which CD82 prevents cancer metastasis is unclear. This study aims to identify genes that are regulated by CD82 in human prostate cell lines. METHODS We used whole human genome microarray to obtain gene expression profiles in a normal prostate epithelial cell line that expressed CD82 (PrEC-31) and a metastatic prostate cell line that does not express CD82 (PC3). Then, siRNA silencing was used to knock down CD82 expression in PrEC-31 while CD82 was re-expressed in PC3 to acquire differentially-expressed genes in the respective cell line. RESULTS Differentially-expressed genes with a P < 0.05 were identified in 3 data sets: PrEC-31 (+CD82) vs PrEC-31(-CD82), PC3-57 (+CD82) vs. PC3-5 V (-CD82), and PC3-29 (+CD82) vs. PC3-5 V (-CD82). Top 25 gene lists did not show overlap within the data sets, except (CALB1) the calcium binding protein calbindin 1 which was significantly up-regulated (2.8 log fold change) in PrEC-31 and PC3-29 cells that expressed CD82. Other most significantly up-regulated genes included serine peptidase inhibitor kazal type 1 (SPINK1) and polypeptide N-acetyl galactosaminyl transferase 14 (GALNT14) and most down-regulated genes included C-X-C motif chemokine ligand 14 (CXCL14), urotensin 2 (UTS2D), and fibroblast growth factor 13 (FGF13). Pathways related with cell proliferation and angiogenesis, migration and invasion, cell death, cell cycle, signal transduction, and metabolism were highly enriched in cells that lack CD82 expression. Expression of two mutually inclusive genes in top 100 gene lists of all data sets, runt-related transcription factor (RUNX3) and trefoil factor 3 (TFF3), could be validated with qRT-PCR. CONCLUSION Identification of genes and pathways regulated by CD82 in this study may provide additional insights into the role that CD82 plays in prostate tumor progression and metastasis, as well as identify potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Pushpaja Dodla
- Department of Cell and Molecular Biology, Grand Valley State University, Allendale, MI, 49401, USA
| | - Vanitha Bhoopalan
- Department of Cell and Molecular Biology, Grand Valley State University, Allendale, MI, 49401, USA
| | - Sok Kean Khoo
- Department of Cell and Molecular Biology, Grand Valley State University, Allendale, MI, 49401, USA
| | - Cindy Miranti
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ, 85724, USA
| | - Suganthi Sridhar
- Department of Integrative Biology, University of South Florida, 140, 7Th Avenue S, University of South Florida, St. Petersburg, FL, 33701, USA.
| |
Collapse
|
38
|
Cao J, O'Day DR, Pliner HA, Kingsley PD, Deng M, Daza RM, Zager MA, Aldinger KA, Blecher-Gonen R, Zhang F, Spielmann M, Palis J, Doherty D, Steemers FJ, Glass IA, Trapnell C, Shendure J. A human cell atlas of fetal gene expression. Science 2020; 370:370/6518/eaba7721. [PMID: 33184181 DOI: 10.1126/science.aba7721] [Citation(s) in RCA: 422] [Impact Index Per Article: 84.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 09/10/2020] [Indexed: 12/14/2022]
Abstract
The gene expression program underlying the specification of human cell types is of fundamental interest. We generated human cell atlases of gene expression and chromatin accessibility in fetal tissues. For gene expression, we applied three-level combinatorial indexing to >110 samples representing 15 organs, ultimately profiling ~4 million single cells. We leveraged the literature and other atlases to identify and annotate hundreds of cell types and subtypes, both within and across tissues. Our analyses focused on organ-specific specializations of broadly distributed cell types (such as blood, endothelial, and epithelial), sites of fetal erythropoiesis (which notably included the adrenal gland), and integration with mouse developmental atlases (such as conserved specification of blood cells). These data represent a rich resource for the exploration of in vivo human gene expression in diverse tissues and cell types.
Collapse
Affiliation(s)
- Junyue Cao
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Diana R O'Day
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Hannah A Pliner
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Paul D Kingsley
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| | - Mei Deng
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Riza M Daza
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Michael A Zager
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.,Center for Data Visualization, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Kimberly A Aldinger
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA.,Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Ronnie Blecher-Gonen
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | | | - Malte Spielmann
- Human Molecular Genomics Group, Max Planck Institute for Molecular Genetics, Berlin, Germany.,Institute of Human Genetics, University of Lübeck, Lübeck, Germany
| | - James Palis
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| | - Dan Doherty
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA.,Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.,Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | | | - Ian A Glass
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA.,Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.,Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Cole Trapnell
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA. .,Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.,Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA. .,Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.,Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA.,Howard Hughes Medical Institute, Seattle, WA, USA
| |
Collapse
|
39
|
Ochiai Y, Yamaguchi J, Kokuryo T, Yokoyama Y, Ebata T, Nagino M. Trefoil Factor Family 1 Inhibits the Development of Hepatocellular Carcinoma by Regulating β-Catenin Activation. Hepatology 2020; 72:503-517. [PMID: 31733149 DOI: 10.1002/hep.31039] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 11/07/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS Recent studies have suggested that trefoil factor family 1 (TFF1) functions as a tumor suppressor in gastric and pancreatic carcinogenesis. APPROACH AND RESULTS To investigate the role of TFF1 in hepatocarcinogenesis, we performed immunohistochemical staining of surgically resected human liver samples, transfected a TFF1 expression vector into hepatocellular carcinoma (HCC) cell lines, and employed a mouse model of spontaneous HCC development (albumin-cyclization recombination/Lox-Stop-Lox sequence-Kirsten rat sarcoma viral oncogene homologG12D [KC]); the model mouse strain was bred with a TFF1-knockout mouse strain to generate a TFF1-deficient HCC mouse model (KC/TFF1-/- ). TFF1 expression was found in some human samples with HCC. Interestingly, TFF1-positive cancer cells showed a staining pattern contradictory to that of proliferating cell nuclear antigen, and aberrant DNA hypermethylation in TFF1 promoter lesions was detected in HCC samples, indicating the tumor-suppressive role of TFF1. In vitro, induction of TFF1 expression resulted in impaired proliferative activity and enhanced apoptosis in HCC cell lines (HuH7, HepG2, and HLE). These anticancer effects of TFF1 were accompanied by the loss of nuclear β-catenin expression, indicating inactivation of the β-catenin signaling pathway by TFF1. In vivo, TFF1 deficiency in KC mice accelerated the early development and growth of HCC, resulting in poor survival rates. In addition, immunohistochemistry revealed that the amount of nuclear-localized β-catenin was significantly higher in KC/TFF1-/- mice than in KC mice and that human HCC tissue showed contradictory expression patterns for β-catenin and TFF1, confirming the in vitro observations. CONCLUSIONS TFF1 might function as a tumor suppressor that inhibits the development of HCC by regulating β-catenin activity.
Collapse
Affiliation(s)
- Yosuke Ochiai
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Junpei Yamaguchi
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toshio Kokuryo
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yukihiro Yokoyama
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomoki Ebata
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masato Nagino
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
40
|
Hoffmann W. Trefoil Factor Family (TFF) Peptides and Their Diverse Molecular Functions in Mucus Barrier Protection and More: Changing the Paradigm. Int J Mol Sci 2020; 21:ijms21124535. [PMID: 32630599 PMCID: PMC7350206 DOI: 10.3390/ijms21124535] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/17/2020] [Accepted: 06/19/2020] [Indexed: 02/07/2023] Open
Abstract
Trefoil factor family peptides (TFF1, TFF2, TFF3) are typically co-secreted together with mucins. Tff1 represents a gastric tumor suppressor gene in mice. TFFs are also synthesized in minute amounts in the immune and central nervous systems. In mucous epithelia, they support rapid repair by enhancing cell migration ("restitution") via their weak chemotactic and anti-apoptotic effects. For a long time, as a paradigm, this was considered as their major biological function. Within recent years, the formation of disulfide-linked heterodimers was documented for TFF1 and TFF3, e.g., with gastrokine-2 and IgG Fc binding protein (FCGBP). Furthermore, lectin activities were recognized as enabling binding to a lipopolysaccharide of Helicobacter pylori (TFF1, TFF3) or to a carbohydrate moiety of the mucin MUC6 (TFF2). Only recently, gastric TFF1 was demonstrated to occur predominantly in monomeric forms with an unusual free thiol group. Thus, a new picture emerged, pointing to diverse molecular functions for TFFs. Monomeric TFF1 might protect the gastric mucosa as a scavenger for extracellular reactive oxygen/nitrogen species. Whereas, the TFF2/MUC6 complex stabilizes the inner layer of the gastric mucus. In contrast, the TFF3-FCGBP heterodimer (and also TFF1-FCGBP) are likely part of the innate immune defense of mucous epithelia, preventing the infiltration of microorganisms.
Collapse
Affiliation(s)
- Werner Hoffmann
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| |
Collapse
|
41
|
Braga Emidio N, Brierley SM, Schroeder CI, Muttenthaler M. Structure, Function, and Therapeutic Potential of the Trefoil Factor Family in the Gastrointestinal Tract. ACS Pharmacol Transl Sci 2020; 3:583-597. [PMID: 32832864 DOI: 10.1021/acsptsci.0c00023] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Indexed: 12/20/2022]
Abstract
Trefoil factor family peptides (TFF1, TFF2, and TFF3) are key players in protecting, maintaining, and repairing the gastrointestinal tract. Accordingly, they have the therapeutic potential to treat and prevent a variety of gastrointestinal disorders associated with mucosal damage. TFF peptides share a conserved motif, including three disulfide bonds that stabilize a well-defined three-loop-structure reminiscent of a trefoil. Although multiple functions have been described for TFF peptides, their mechanisms at the molecular level remain poorly understood. This review presents the status quo of TFF research relating to gastrointestinal disorders. Putative TFF receptors and protein partners are described and critically evaluated. The therapeutic potential of these peptides in gastrointestinal disorders where altered mucosal biology plays a crucial role in the underlying etiology is discussed. Finally, areas of investigation that require further research are addressed. Thus, this review provides a comprehensive update on TFF literature as well as guidance toward future research to better understand this peptide family and its therapeutic potential for the treatment of gastrointestinal disorders.
Collapse
Affiliation(s)
- Nayara Braga Emidio
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Stuart M Brierley
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medicial Research Insittitue (FHMRI), Flinders University, Bedford Park, South Australia 5042, Australia.,Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia 5000, Australia.,Discipline of Medicine, University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Christina I Schroeder
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia.,National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702, United States
| | - Markus Muttenthaler
- Institute of Biological Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria.,Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
42
|
Jahan R, Shah A, Kisling SG, Macha MA, Thayer S, Batra SK, Kaur S. Odyssey of trefoil factors in cancer: Diagnostic and therapeutic implications. Biochim Biophys Acta Rev Cancer 2020; 1873:188362. [PMID: 32298747 DOI: 10.1016/j.bbcan.2020.188362] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/01/2020] [Accepted: 04/01/2020] [Indexed: 02/07/2023]
Abstract
Trefoil factors 1, 2, and 3 (TFFs) are a family of small secretory molecules involved in the protection and repair of the gastrointestinal tract (GI). TFFs maintain and restore epithelial structural integrity via transducing key signaling pathways for epithelial cell migration, proliferation, and invasion. In recent years, TFFs have emerged as key players in the pathogenesis of multiple diseases, especially cancer. Initially recognized as tumor suppressors, emerging evidence demonstrates their key role in tumor progression and metastasis, extending their actions beyond protection. However, to date, a comprehensive understanding of TFFs' mechanism of action in tumor initiation, progression and metastasis remains obscure. The present review discusses the structural, functional and mechanistic implications of all three TFF family members in tumor progression and metastasis. Also, we have garnered information from studies on their structure and expression status in different organs, along with lessons from their specific knockout in mouse models. In addition, we highlight the emerging potential of using TFFs as a biomarker to stratify tumors for better therapeutic intervention.
Collapse
Affiliation(s)
- Rahat Jahan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, NE, 68198, USA
| | - Ashu Shah
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, NE, 68198, USA
| | - Sophia G Kisling
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, NE, 68198, USA
| | - Muzafar A Macha
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, NE, 68198, USA; Department of Otolaryngology-Head & Neck Surgery, University of Nebraska Medical Center, NE, 68198, USA; Department of Biotechnology, Central University of Kashmir, Ganderbal, Jammu and Kashmir, India -191201
| | - Sarah Thayer
- Division of Surgical Oncology, Department of Surgery, University of Nebraska Medical Center, NE, 68198, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, NE, 68198, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, NE, 68198, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, NE, 68198, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, NE 68198, USA.
| | - Sukhwinder Kaur
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, NE, 68198, USA.
| |
Collapse
|
43
|
Yi J, Ren L, Li D, Wu J, Li W, Du G, Wang J. Trefoil factor 1 (TFF1) is a potential prognostic biomarker with functional significance in breast cancers. Biomed Pharmacother 2020; 124:109827. [PMID: 31986408 DOI: 10.1016/j.biopha.2020.109827] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/07/2019] [Accepted: 12/13/2019] [Indexed: 01/25/2023] Open
Abstract
Breast cancer (BC) is the most common cancer in women and the second leading cause of their cancer death. Establishing an accurate BC prognosis is very difficult because of its heterogeneity. Elevated TFF1 levels in serum were associated with development of BC, TFF1 expression was upregulated in BC compared to the healthy breast tissue. The aim of this study was to investigate the function of TFF1 in BCs, and to assess whether serum TFF1 could be used in formulating a prognosis for BC patients. In silico analyses were carried out to determine the expression of TFF1 mRNA in different types of BC and the association between TFF1 expression and survival of BC patients. Expression of TFF1 protein was checked in 52 paraffin-embedded tissues of BCs by immunochemistry, and serum concentration of TFF1 in 70 BC patients and 32 healthy controls was measured by ELISA. Functional activities of TFF1 in BC cells were determined by CCK-8 assay, colony formation, BrdU-DNA synthesis, and assays for migration and invasion. Results showed that expression of TFF1 mRNA was correlated with expression of biomarkers of luminal cancers including ESR1, GATA3, FOXA1, MYB and XBP1. In addition, patients with ER+BC had higher expression of TFF1 than those with ER- (p < 0.05). There was also lower expression of TFF1 in triple-negative breast cancer (TNBC) than in non-TNBC (p < 0.05), which corresponds with the level of serum TFF1 in TNBC patients, compared with non-TNBC patients (p < 0.001). Furthermore, expression of TFF1 was associated with tumor size (p = 0.002), nodal status (p < 0.001), histological grade (p < 0.001), ER status (p = 0.012), PR status (p < 0.001) and HER2 (p < 0.001), while serum TFF1 was only statistically different among BC with ER+, PR + and HER2+ (p = 0.04139, 0.0018, 0.0004). Elevated TFF1 expression correlated with increased overall survival of BC patients (p = 0.00068). Finally, TFF1 was found to inhibit the cell growth, colony formation, migration and invasion of BC cells in vitro. All these results suggest that expression of TFF1 was related to ER status of BC and that expression of TFF1 was lower in TNBC than in non-TNBC. TFF1 was found to inhibit proliferation, migration and invasion of BC cells in vitro. Expression of TFF1 was associated with clinical characters of patients with BC. Serum TFF1 could be used to predict prognosis of patients with BC, especially non-TNBC.
Collapse
Affiliation(s)
- Jie Yi
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Liwen Ren
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Dandan Li
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Jie Wu
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Wan Li
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Guanhua Du
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Jinhua Wang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China.
| |
Collapse
|
44
|
Popp J, Schicht M, Garreis F, Klinger P, Gelse K, Sesselmann S, Tsokos M, Etzold S, Stiller D, Claassen H, Paulsen F. Human Synovia Contains Trefoil Factor Family (TFF) Peptides 1-3 Although Synovial Membrane Only Produces TFF3: Implications in Osteoarthritis and Rheumatoid Arthritis. Int J Mol Sci 2019; 20:ijms20236105. [PMID: 31817054 PMCID: PMC6928748 DOI: 10.3390/ijms20236105] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 11/27/2019] [Accepted: 11/28/2019] [Indexed: 01/15/2023] Open
Abstract
Objective: Trefoil factor family peptide 3 (TFF3) has been shown to support catabolic functions in cases of osteoarthritis (OA). As in joint physiology and diseases such as OA, the synovial membrane (SM) of the joint capsule also plays a central role. We analyze the ability of SM to produce TFF compare healthy SM and its secretion product synovial fluid (SF) with SM and SF from patients suffering from OA or rheumatoid arthritis (RA). Methods: Real-time PCR and ELISA were used to measure the expression of TFFs in healthy SM and SM from patients suffering from OA or RA. For tissue localization, we investigated TFF1-3 in differently aged human SM of healthy donors by means of immunohistochemistry, real-time PCR and Western blot. Results: Only TFF3 but not TFF1 and -2 was expressed in SM from healthy donors as well as cases of OA or RA on protein and mRNA level. In contrast, all three TFFs were detected in all samples of SF on the protein level. No significant changes were observed for TFF1 at all. TFF2 was significantly upregulated in RA samples in comparison to OA samples. TFF3 protein was significantly downregulated in OA samples in comparison to healthy samples and cases of RA significantly upregulated compared to OA. In contrast, in SM TFF3 protein was not significantly regulated. Conclusion: The data demonstrate the production of TFF3 in SM. Unexpectedly, SF contains all three known TFF peptides. As neither articular cartilage nor SM produce TFF1 and TFF2, we speculate that these originate with high probability from blood serum.
Collapse
Affiliation(s)
- Judith Popp
- Friedrich Alexander University Erlangen-Nürnberg (FAU), Institute of Functional and Clinical Anatomy, 91054 Erlangen, Germany; (J.P.); (M.S.); (F.G.); (P.K.)
| | - Martin Schicht
- Friedrich Alexander University Erlangen-Nürnberg (FAU), Institute of Functional and Clinical Anatomy, 91054 Erlangen, Germany; (J.P.); (M.S.); (F.G.); (P.K.)
| | - Fabian Garreis
- Friedrich Alexander University Erlangen-Nürnberg (FAU), Institute of Functional and Clinical Anatomy, 91054 Erlangen, Germany; (J.P.); (M.S.); (F.G.); (P.K.)
| | - Patricia Klinger
- Friedrich Alexander University Erlangen-Nürnberg (FAU), Institute of Functional and Clinical Anatomy, 91054 Erlangen, Germany; (J.P.); (M.S.); (F.G.); (P.K.)
| | - Kolja Gelse
- University Hospital Erlangen, Department of Trauma Surgery, 91054 Erlangen, Germany;
| | - Stefan Sesselmann
- University of Applied Sciences Amberg-Weiden, Institute for Medical Engineering, 92637 Weiden, Germany;
| | - Michael Tsokos
- Charité-Universitätsmedizin Berlin, Institute of Legal Medicine and Forensic Sciences, 10117 Berlin, Germany; (M.T.); (S.E.)
| | - Saskia Etzold
- Charité-Universitätsmedizin Berlin, Institute of Legal Medicine and Forensic Sciences, 10117 Berlin, Germany; (M.T.); (S.E.)
| | - Dankwart Stiller
- Martin Luther University Halle-Wittenberg (MLU), Department of Legal Medicine, 06108 Halle (Saale), Germany;
| | - Horst Claassen
- Martin Luther University Halle-Wittenberg (MLU), Department of Anatomy and Cell Biology, 06108 Halle (Saale), Germany;
| | - Friedrich Paulsen
- Friedrich Alexander University Erlangen-Nürnberg (FAU), Institute of Functional and Clinical Anatomy, 91054 Erlangen, Germany; (J.P.); (M.S.); (F.G.); (P.K.)
- Sechenov University, Department of Topographic Anatomy and Operative Surgery, 119146 Moscow, Russia
- Correspondence: ; Tel.: +49-9131-8522865; Fax: +49-9131-8522862
| |
Collapse
|
45
|
Mihalj M, Bujak M, Butković J, Zubčić Ž, Tolušić Levak M, Čes J, Kopić V, Baus Lončar M, Mihalj H. Differential Expression of TFF1 and TFF3 in Patients Suffering from Chronic Rhinosinusitis with Nasal Polyposis. Int J Mol Sci 2019; 20:ijms20215461. [PMID: 31683988 PMCID: PMC6862153 DOI: 10.3390/ijms20215461] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 10/21/2019] [Accepted: 10/30/2019] [Indexed: 12/18/2022] Open
Abstract
Trefoil family factor (TFF) proteins contribute to antimicrobial defense and the maintenance of sinonasal epithelial barrier integrity. Dysregulation of TFF expression may be involved in the development of chronic inflammation and tissue remodeling characteristically found in chronic rhinosinusitis with nasal polyposis (CRSwNP). Expressions of TFF1 and TFF3 were determined in specimens of middle nasal turbinate (MNT-0), bulla ethmoidalis (BE), and nasal polyps (NP) from CRSwNP patients (n = 29) and inferior nasal turbinate from a group of control patients (underwent nasal septoplasty, n = 25). An additional MNT sample was collected 6 months after functional endoscopic sinus surgery (FESS, MNT-6). TFF1 mRNA levels were significantly reduced in all specimens by approximately three- to five-fold, while TFF3 was increased in MNT-0, as compared with controls. Six months after surgery their levels were reversed to control values. CRSwNP patients with S. epidermidis isolated from sinus swabs showed upregulation of TFF3 in MNT and NP as compared with patients with sterile swabs. Target gene regulation was not affected by the presence of type 2 inflammation in patients with confirmed allergy. Results of this study imply participation of TFFs genes in the development of CRSwNP.
Collapse
Affiliation(s)
- Martina Mihalj
- Department of Dermatology and Venereology, University Hospital Osijek, 31000 Osijek, Croatia.
- Department of Physiology and Immunology, Faculty of Medicine, University of Osijek, 31000 Osijek, Croatia.
| | - Maro Bujak
- Department of Materials Chemistry, Ruđer Bošković Institute, 10000 Zagreb, Croatia.
| | - Josip Butković
- Department of Maxillofacial Surgery, University Hospital Osijek, 310000 Osijek, Croatia.
- Department of Otorhinolaryngology and Maxillofacial Surgery, Faculty of Medicine, University of Osijek, 31000 Osijek, Croatia.
| | - Željko Zubčić
- Department of Otorhinolaryngology and Maxillofacial Surgery, Faculty of Medicine, University of Osijek, 31000 Osijek, Croatia.
- Department of Otorhinolaryngology and Head and Neck Surgery, University Hospital Osijek; 31000 Osijek, Croatia.
| | - Maja Tolušić Levak
- Department of Dermatology and Venereology, University Hospital Osijek, 31000 Osijek, Croatia.
- Department of Histology and Embryology, Faculty of Medicine, University of Osijek, 31000 Osijek, Croatia.
| | - Josip Čes
- Dental Centre Čes, 31000 Osijek, Croatia.
| | - Vlatko Kopić
- Department of Maxillofacial Surgery, University Hospital Osijek, 310000 Osijek, Croatia.
- Department of Otorhinolaryngology and Maxillofacial Surgery, Faculty of Medicine, University of Osijek, 31000 Osijek, Croatia.
| | - Mirela Baus Lončar
- Department of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia.
| | - Hrvoje Mihalj
- Department of Otorhinolaryngology and Maxillofacial Surgery, Faculty of Medicine, University of Osijek, 31000 Osijek, Croatia.
- Department of Otorhinolaryngology and Head and Neck Surgery, University Hospital Osijek; 31000 Osijek, Croatia.
| |
Collapse
|
46
|
The Interaction of Helicobacter pylori with TFF1 and Its Role in Mediating the Tropism of the Bacteria Within the Stomach. Int J Mol Sci 2019; 20:ijms20184400. [PMID: 31500233 PMCID: PMC6769565 DOI: 10.3390/ijms20184400] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 08/27/2019] [Accepted: 09/06/2019] [Indexed: 02/07/2023] Open
Abstract
Helicobacter pylori colonises the human stomach and has tropism for the gastric mucin, MUC5AC. The majority of organisms live in the adherent mucus layer within their preferred location, close to the epithelial surface where the pH is near neutral. Trefoil factor 1 (TFF1) is a small trefoil protein co-expressed with the gastric mucin MUC5AC in surface foveolar cells and co-secreted with MUC5AC into gastric mucus. Helicobacter pylori binds with greater avidity to TFF1 dimer, which is present in gastric mucus, than to TFF1 monomer. Binding of H. pylori to TFF1 is mediated by the core oligosaccharide subunit of H. pylori lipopolysaccharide at pH 5.0–6.0. Treatment of H. pylori lipopolysaccharide with mannosidase or glucosidase inhibits its interaction with TFF1. Both TFF1 and H. pylori have a propensity for binding to mucins with terminal non-reducing α- or β-linked N-acetyl-d-glucosamine or α-(2,3) linked sialic acid or Gal-3-SO42−. These findings are strong evidence that TFF1 has carbohydrate-binding properties that may involve a conserved patch of aromatic hydrophobic residues on the surface of its trefoil domain. The pH-dependent lectin properties of TFF1 may serve to locate H. pylori deep in the gastric mucus layer close to the epithelium rather than at the epithelial surface. This restricted localisation could limit the interaction of H. pylori with epithelial cells and the subsequent host signalling events that promote inflammation.
Collapse
|
47
|
Hormdee D, Prajaneh S, Kampichai A, Tak R, Chaiyarit P. Prolonged Suppressive Effects of Periodontitis on Salivary TFF3 Production. Eur J Dent 2019; 13:193-198. [PMID: 31466117 PMCID: PMC6777149 DOI: 10.1055/s-0039-1693949] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Objective
As a follow-up to our previous study that demonstrated decreased salivary trefoil factor family 3 (TFF3) peptide levels in chronic periodontitis patients, this current study aimed to observe the effects of nonsurgical periodontal treatment on salivary TFF3 peptides in patients with periodontal diseases.
Materials and Methods
Eighty-seven volunteers that comprised of 30 individuals with healthy periodontium, 31 with gingivitis, and 26 with chronic periodontitis were considered for the study. Prior to periodontal treatment, a general periodontal examination was performed along with collection of saliva samples from each volunteer. Nonsurgical periodontal treatments were provided to patients with gingivitis and periodontitis. Two weeks post-treatment, saliva samples were recollected, and the periodontal status was re-evaluated. Salivary TFF3 concentrations were measured by enzyme-linked immunosorbent assay.
Statistical Analysis
Mann–Whitney U test was used when the investigated data were not normally distributed. Chi-squared test was used when dealing with categorical data. Kruskal–Wallis test with post-hoc corrections was used to compare data among the three investigated groups. Two-tailed
p
< 0.05 was considered as statistically significant.
Results
Prior to the periodontal treatment, salivary TFF3 concentrations in patients with gingivitis and periodontitis were significantly lower than those with healthy periodontium. Two weeks post-treatment, increased levels of salivary TFF3 were observed in patients with gingivitis, whereas the concentrations decreased in patients with chronic periodontitis.
Conclusion
This study demonstrated the effects of periodontal disease on the production of salivary TFF3 peptides. Interestingly, nonsurgical periodontal treatment also affected the recovery of salivary TFF3 peptides but varied in their outcomes between gingivitis and periodontitis patients.
Collapse
Affiliation(s)
- Doosadee Hormdee
- Division of Periodontology, Department of Oral Biomedical Sciences, Faculty of Dentistry, Khon Kaen University, Khon Kaen, Thailand.,Research Group of Chronic Inflammatory Oral Diseases and Systemic Diseases Associated with Oral Health, Khon Kaen University, Thailand
| | - Saengsome Prajaneh
- Division of Periodontology, Department of Oral Biomedical Sciences, Faculty of Dentistry, Khon Kaen University, Khon Kaen, Thailand
| | | | - Ranuch Tak
- Faculty of Dentistry, International University, Phnom Penh, Cambodia
| | - Ponlatham Chaiyarit
- Research Group of Chronic Inflammatory Oral Diseases and Systemic Diseases Associated with Oral Health, Khon Kaen University, Thailand.,Division of Oral Diagnosis, Department of Oral Biomedical Sciences, Faculty of Dentistry, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
48
|
Yusufu A, Shayimu P, Tuerdi R, Fang C, Wang F, Wang H. TFF3 and TFF1 expression levels are elevated in colorectal cancer and promote the malignant behavior of colon cancer by activating the EMT process. Int J Oncol 2019; 55:789-804. [PMID: 31432157 PMCID: PMC6741840 DOI: 10.3892/ijo.2019.4854] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 06/27/2019] [Indexed: 12/19/2022] Open
Abstract
Reports on the roles of the secreted trefoil factor (TFF)1 and 3 in colorectal cancer (CRC) and their underlying mechanisms of action in tumorigenesis are not common and are controversial. In the present study, the mRNA expression and promoter methylation of TFF1 and TFF3 in cancer and adjacent normal tissues were investigated, and their association with other clinical factors and patient prognosis were evaluated. Moreover, the association between TFF3 and epithelial mesenchymal transition (EMT) was explored by overexpressing or inhibiting TFF3 expression. The results revealed that the mRNA level of TFF1 and TFF3 in the cancer tissues was significantly higher than that in the matched adjacent normal tissues (P=0.034 and P=0.007, respectively), and a higher expression of TFF3, but not TFF1, was predominantly associated with clinicopathological factors and a poorer prognosis. No correlation was observed between promoter methylation and the expression of TFF1 or TFF3. The overexpression of TFF3 promoted the proliferation, migration and invasiveness of HT29 cells, and induced an increase in the expression of Twist1, Snail and Vimentin, while causing a decrease in E-cadherin expression. On the contrary, the knockdown of TFF3 resulted in opposite effects in the LoVo cells. On the whole, the findings of this study indicate that TFF3 may be a promising new factor for the estimation of the survival of patients with CRC, and may promote the malignant progression of CRC by activating the EMT process. Therefore, TFF3 may be a future potential therapeutic target for CRC.
Collapse
Affiliation(s)
- Aikeremu Yusufu
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital, Xin Jiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830011, P.R. China
| | - Paerhati Shayimu
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital, Xin Jiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830011, P.R. China
| | - Rousidan Tuerdi
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital, Xin Jiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830011, P.R. China
| | - Cheng Fang
- Department of Gastrointestinal Surgery, Xi Jing Digestive Disease Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Fei Wang
- Department of Gastrointestinal Surgery, Xi Jing Digestive Disease Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Haijiang Wang
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital, Xin Jiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830011, P.R. China
| |
Collapse
|
49
|
Wang Y, Kim R, Sims CE, Allbritton NL. Building a Thick Mucus Hydrogel Layer to Improve the Physiological Relevance of In Vitro Primary Colonic Epithelial Models. Cell Mol Gastroenterol Hepatol 2019; 8:653-655.e5. [PMID: 31356887 PMCID: PMC6889783 DOI: 10.1016/j.jcmgh.2019.07.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 07/18/2019] [Accepted: 07/19/2019] [Indexed: 02/07/2023]
Affiliation(s)
- Y Wang
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - R Kim
- UNC/NC State Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
| | - C E Sims
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - N L Allbritton
- UNC/NC State Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, and North Carolina State University, Raleigh, North Carolina.
| |
Collapse
|
50
|
Role of lysophosphatidic acid in proliferation and differentiation of intestinal epithelial cells. PLoS One 2019; 14:e0215255. [PMID: 31017922 PMCID: PMC6481811 DOI: 10.1371/journal.pone.0215255] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 03/28/2019] [Indexed: 11/19/2022] Open
Abstract
Intestinal epithelial cells (IECs) are regenerated continuously from intestinal stem cells (ISCs) near the base of intestinal crypts in order to maintain homeostasis and structural integrity of intestinal epithelium. Epidermal growth factor (EGF) is thought to be important to drive the proliferation and differentiation of IECs from ISCs, it remains unknown whether other growth factors or lipid mediators are also important for such regulation, however. Here we show that lysophosphatidic acid (LPA), instead of EGF, robustly promoted the development of intestinal organoids prepared from the mouse small intestine. Indeed, LPA exhibited the proliferative activity of IECs as well as induction of differentiation of IECs into goblet cells, Paneth cells, and enteroendocrine cells in intestinal organoids. Inhibitors for LPA receptor 1 markedly suppressed the LPA-promoted development of intestinal organoids. LPA also promoted the phosphorylation of extracellular signal-regulated kinase (ERK) 1/2 in intestinal organoids, whereas inhibition of mitogen-activated protein kinase/ERK kinase (MEK) 1/2 significantly suppressed the development of, as well as the proliferative activity and differentiation of, intestinal organoids in response to LPA. Our results thus suggest that LPA is a key factor that drives the proliferation and differentiation of IECs.
Collapse
|