Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 11(1): 15830, 2021 08 04.
Article in English | MEDLINE | ID: mdl-34349157

ABSTRACT

The heart is capable of activating protective mechanisms in response to ischemic injury to support myocardial survival and performance. These mechanisms have been recognized primarily in the ischemic heart, involving paracrine signaling processes. Here, we report a distant cardioprotective mechanism involving hepatic cell mobilization to the ischemic myocardium in response to experimental myocardial ischemia-reperfusion (MI-R) injury. A parabiotic mouse model was generated by surgical skin-union of two mice and used to induce bilateral MI-R injury with unilateral hepatectomy, establishing concurrent gain- and loss-of-hepatic cell mobilization conditions. Hepatic cells, identified based on the cell-specific expression of enhanced YFP, were found in the ischemic myocardium of parabiotic mice with intact liver (0.2 ± 0.1%, 1.1 ± 0.3%, 2.7 ± 0.6, and 0.7 ± 0.4% at 1, 3, 5, and 10 days, respectively, in reference to the total cell nuclei), but not significantly in the ischemic myocardium of parabiotic mice with hepatectomy (0 ± 0%, 0.1 ± 0.1%, 0.3 ± 0.2%, and 0.08 ± 0.08% at the same time points). The mobilized hepatic cells were able to express and release trefoil factor 3 (TFF3), a protein mitigating MI-R injury as demonstrated in TFF3-/- mice (myocardium infarcts 17.6 ± 2.3%, 20.7 ± 2.6%, and 15.3 ± 3.8% at 1, 5, and 10 days, respectively) in reference to wildtype mice (11.7 ± 1.9%, 13.8 ± 2.3%, and 11.0 ± 1.8% at the same time points). These observations suggest that MI-R injury can induce hepatic cell mobilization to support myocardial survival by releasing TFF3.


Subject(s)
Cardiotonic Agents/metabolism , Disease Models, Animal , Liver Transplantation/methods , Liver/metabolism , Myocardial Reperfusion Injury/prevention & control , Trefoil Factor-3/physiology , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology
3.
Int J Mol Sci ; 20(17)2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31480518

ABSTRACT

The aim of the study was to explore the possible role of Trefoil Factor Family peptide 3 (TFF3) for skeletal repair. The expression of TFF3 was analyzed in human joint tissues as well as in a murine bone fracture model. Serum levels of TFF3 following a defined skeletal trauma in humans were determined by ELISA. The mRNA expression of TFF3 was analyzed under normoxia and hypoxia. Expression analysis after stimulation of human mesenchymal progenitor cells (MPCs) with TFF3 was performed by RT2 Profiler PCR Array. The effect of recombinant human (rh)TFF3 on MPCs was analysed by different migration and chemotaxis assays. The effect on cell motility was also visualized by fluorescence staining of F-Actin. TFF3 was absent in human articular cartilage, but strongly expressed in the subchondral bone and periosteum of adult joints. Strong TFF3 immunoreactivity was also detected in murine fracture callus. Serum levels of TFF3 were significantly increased after skeletal trauma in humans. Expression analysis demonstrated that rhTFF3 significantly decreased mRNA of ROCK1. Wound healing assays showed increased cell migration of MPCs by rhTFF3. The F-Actin cytoskeleton was markedly influenced by rhTFF3. Cell proliferation was not increased by rhTFF3. The data demonstrate elevated expression of TFF3 after skeletal trauma. The stimulatory effects on cell motility and migration of MPCs suggest a role of TFF3 in skeletal repair.


Subject(s)
Actin Cytoskeleton/metabolism , Bone and Bones/physiology , Cell Movement , Trefoil Factor-3/metabolism , Aged , Aged, 80 and over , Animals , Bone and Bones/metabolism , Female , Fracture Healing , Gene Expression Regulation , Humans , Hypoxia , Mice , Mice, Inbred C57BL , Middle Aged , Trefoil Factor-3/physiology , rho-Associated Kinases/genetics
4.
World J Surg ; 42(12): 3997-4004, 2018 12.
Article in English | MEDLINE | ID: mdl-30039286

ABSTRACT

BACKGROUND: Trefoil factor 3 (TFF3) is a small molecule secreted by the mammalian gastrointestinal tract and is overexpressed in some human malignant tumors. We investigated the prognostic values of immunohistochemical (IHC) TFF3 expression and serum TFF3 levels in patients with gastric cancer, and whether TFF3 influenced tumor proliferation and invasion in vitro. METHODS: We examined 111 patients who underwent R0 gastrectomy for gastric cancer between April 2012 and April 2015. IHC TFF3 expression and serum TFF3 levels were evaluated regarding their associations with clinicopathological factors and recurrence-free survival (RFS). In vitro cell proliferation and migration assays were used to explore the biological role of TFF3 in human gastric cancer cell lines following transfection with a lentivirus-based shRNA plasmid. RESULTS: IHC TFF3 expression showed significant associations with depth of invasion (p = 0.024), lymph node metastasis (p = 0.008), and RFS (log-rank p = 0.002). Serum TFF3 levels were correlated with IHC TFF3 expression (p = 0.013). RFS was significantly poorer in patients with high (n = 27) compared to low (n = 84) serum TFF3 levels (log-rank p = 0.003). Cox multivariate analysis indicated that serum TFF3 level was an independent prognostic factor for RFS (p = 0.024). In vitro assays, TFF3 downregulation significantly inhibited both proliferation and invasion of gastric cancer cells. CONCLUSIONS: Serum TFF3 levels could be useful prognostic markers in patients with gastric cancer. TFF3 may play various biological roles in proliferation and invasion of gastric cancer cells.


Subject(s)
Stomach Neoplasms/mortality , Trefoil Factor-3/blood , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Female , Humans , Immunohistochemistry , Lymphatic Metastasis , Male , Middle Aged , Prognosis , Stomach Neoplasms/pathology , Stomach Neoplasms/surgery , Trefoil Factor-3/physiology
5.
Arab J Gastroenterol ; 18(4): 191-200, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29241727

ABSTRACT

BACKGROUND AND STUDY AIM: Colorectal cancer (CRC) is a heterogeneous disease entity with a diverse biological pathogenesis. This study aims to validate the two studies published in 2013 which established a separate CRC molecular subtype classification by utilizing a rapidly accessible miniclassifier, and verify a simplified version thereof. PATIENTS AND METHODS: Participants diagnosed with CRC (n = 568) were subtyped in three classifications for characteristic, and prognostic purposes. Colorectal cancer subtypes (CCS) were classified as: i) CCS1 (CDX2+, microsatellite stable (MSS)/microsatellite instability (MSI)-low), ii) CCS2 (MSI-high), and iii) CCS3 (FRMD6/ZEB1/HTR2B +, CDX2-, MSS/MSI-low]. Simplified CCS (SiCCS) subtypes were grouped as: i) CDX2 (CDX2+, MSS/MSI-low, ZEB1 ≤ 2), ii) MSI-H (MSI-high, CDX2/FRMD6/ZEB1/HTR2B +/-), and iii) ZEB1 (ZEB1 ≥ 2, CDX2-, MSS/MSI-low). New molecular classification (NMC) subtypes were defined as: i) enterocyte (E-C) (MUC2 +), ii) goblet-like (G-L) (MUC2 + and TFF3 +), iii) transit-amplifying (T-A) (CFTR +), and iv) stem-like (S-L) (ZEB1 +). RESULTS: In total, 53.5% (n = 304) CCS, 58.3% (n = 331) SiCCS, and 37.7% (n = 214) NMC tumours could be evaluated. CCS2 and MSI-H CRCs had the most favourable survival outcome, whereas the CCS3, ZEB1 and S-L subtypes showed the poorest prognosis. A significant overlap between CCS3, ZEB1, and S-L tumours was demonstrated. CONCLUSION: There is still a need for a consensus gene expression-based subtyping classification system for CRCs, thereby allowing the categorization of most CRC tumours. This study reveals that a simple and rapidly accessible process could replace the complicated, costly and mostly inapproachable methods clinical practices that have been introduced in the majority of previous studies.


Subject(s)
CDX2 Transcription Factor/physiology , Colorectal Neoplasms/classification , Colorectal Neoplasms/genetics , Cytoskeletal Proteins/physiology , Membrane Proteins/physiology , Receptor, Serotonin, 5-HT2B/physiology , Zinc Finger E-box-Binding Homeobox 1/physiology , Adolescent , Adult , Aged , Aged, 80 and over , Colorectal Neoplasms/pathology , Female , Humans , Male , Microsatellite Instability , Middle Aged , Mucin-2/physiology , Reproducibility of Results , Retrospective Studies , Trefoil Factor-3/physiology , Young Adult
6.
PLoS One ; 11(9): e0163025, 2016.
Article in English | MEDLINE | ID: mdl-27626280

ABSTRACT

Trefoil factor family (TFF) peptides have been shown to effect cell proliferation, apoptosis, migration and invasion of normal cells and various cancer cell lines. In the literature TFF peptides are controversially discussed as tumor suppressors and potential tumor progression factors. In the study presented, we investigated the effect of TFF3 overexpression on growth, viability, migration and tumorigenicity of the human retinoblastoma cell lines Y-79, WERI-Rb1, RBL-13 and RBL-15. As revealed by WST-1 and TUNEL assays as well as DAPI and BrdU cell counts, recombinant human TFF3 significantly lowers retinoblastoma cell viability and increases apoptosis levels. Transient TFF3 overexpression likewise significantly increases RB cell apoptosis. Stable, lentiviral TFF3 overexpression lowers retinoblastoma cell viability, proliferation and growth and significantly increases cell death in retinoblastoma cells. Blockage experiments using a broad-spectrum caspase inhibitor and capase-3 immunocytochemistry revealed the involvement of caspases in general and of caspase-3 in particular in TFF3 induced apoptosis in retinoblastoma cell lines. Soft agarose and in ovo chicken chorioallantoic membrane (CAM) assays revealed that TFF3 overexpression influences anchorage independent growth and significantly decreases the size of tumors forming from retinoblastoma cells. Our study demonstrates that forced TFF3 expression exerts a significant pro-apoptotic, anti-proliferative, and tumor suppressive effect in retinoblastoma cells, setting a starting point for new additive chemotherapeutic approaches in the treatment of retinoblastoma.


Subject(s)
Retinal Neoplasms/metabolism , Retinoblastoma/metabolism , Trefoil Factor-3/physiology , Apoptosis , Blotting, Western , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Cell Survival/physiology , Humans , Real-Time Polymerase Chain Reaction , Retinal Neoplasms/physiopathology , Retinoblastoma/physiopathology , Trefoil Factor-3/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...