1
|
Song L, Kostas M, Laerdahl JK, Skálová M, Janská T, Juzeniene A, Ræstad S, Krivokapic A, Kalantzopoulos GN, Soltes J, Vlk M, Kozempel J, Hassfjell S, Wesche J. Preparation and Characterization of an Engineered FGF1 Conjugated to 161Tb for Targeting of FGFRs. ACS OMEGA 2025; 10:5730-5743. [PMID: 39989790 PMCID: PMC11840634 DOI: 10.1021/acsomega.4c09179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/22/2025] [Accepted: 01/30/2025] [Indexed: 02/25/2025]
Abstract
The fibroblast growth factor receptor family members, FGFR1-4, are frequently overexpressed in various solid tumors, including breast cancer and sarcomas. This overexpression highlights the potential of the family of FGFRs as promising targets for cancer therapy. However, conventional FGFR kinase inhibitors often encounter challenges such as limited efficacy or drug resistance. In this study, we pursue an alternative strategy by designing a conjugate of the FGFR ligand FGF1 with the radioisotope 161Tb, for targeted therapy in FGFR-overexpressing cancer cells. FGF1 was engineered (eFGF1) to incorporate a single cysteine at the C terminus for site-specific labeling with a DOTA chelator. eFGF1-DOTA was mixed with the radioisotope 161Tb under mild conditions, resulting in a labeling efficiency above 90%. The nonradioactive ligands were characterized by mass spectrometry, while radioligands were characterized by thin-layer chromatography. The targeting function of the radioligands was assessed through confocal microscopy, flow cytometry, and Western blot analysis, focusing on binding to cancer cells and the activation of downstream signaling pathways related to FGFR. When compared to MCF-7 and RD cell lines with low FGFR expression, eFGF1-DOTA-Tb[161Tb] radioligands demonstrated significantly higher accumulation in FGFR-overexpressing cell lines (MCF-7 FGFR1 and RMS559), leading to enhanced cytotoxicity. Besides radionuclides, eFGF1 can also deliver doxorubicin (DOX) into cancer cells. Considering these characteristics, eFGF1-DOTA-Tb[161Tb] and eFGF1-DOX emerge as promising candidates for FGFR-targeted cancer therapy, and further evaluation in vivo is warranted.
Collapse
Affiliation(s)
- Linlin Song
- Department
of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, Oslo 0379, Norway
- Centre
for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty
of Medicine, University of Oslo, Montebello, Oslo 0379, Norway
| | - Michal Kostas
- Department
of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, Oslo 0379, Norway
- Centre
for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty
of Medicine, University of Oslo, Montebello, Oslo 0379, Norway
| | - Jon K. Laerdahl
- Department
of Microbiology, Oslo University Hospital,
Rikshospitalet, Oslo 0424, Norway
- ELIXIR
Norway, Department of Informatics, University
of Oslo, Oslo 0316, Norway
| | - Marie Skálová
- Faculty
of Nuclear Sciences and Physical Engineering, Czech Technical University in Prague, Břehová 7, Prague 1 110 00, Czech Republic
| | - Tereza Janská
- Faculty
of Nuclear Sciences and Physical Engineering, Czech Technical University in Prague, Břehová 7, Prague 1 110 00, Czech Republic
| | - Asta Juzeniene
- Department
of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Montebello, Oslo, 0379 Norway
| | - Svein Ræstad
- Department
of Tracer Technology, Institute of Energy
Technology, Instituttveien
18, Kjeller 2007, Norway
| | - Alexander Krivokapic
- Department
of Tracer Technology, Institute of Energy
Technology, Instituttveien
18, Kjeller 2007, Norway
| | - Georgios N. Kalantzopoulos
- Department
of Tracer Technology, Institute of Energy
Technology, Instituttveien
18, Kjeller 2007, Norway
| | - Jaroslav Soltes
- Centrum
výzkumu Řež s.r.o., Hlavní 130, Řež, Husinec 250 68, Czech Republic
| | - Martin Vlk
- Faculty
of Nuclear Sciences and Physical Engineering, Czech Technical University in Prague, Břehová 7, Prague 1 110 00, Czech Republic
| | - Jan Kozempel
- Faculty
of Nuclear Sciences and Physical Engineering, Czech Technical University in Prague, Břehová 7, Prague 1 110 00, Czech Republic
| | | | - Jørgen Wesche
- Department
of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, Oslo 0379, Norway
- Centre
for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty
of Medicine, University of Oslo, Montebello, Oslo 0379, Norway
- Department
of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo 0372, Norway
| |
Collapse
|
2
|
Easter M, Hirsch MJ, Harris E, Howze PH, Matthews EL, Jones LI, Bollenbecker S, Vang S, Tyrrell DJ, Sanders YY, Birket SE, Barnes JW, Krick S. FGF receptors mediate cellular senescence in the cystic fibrosis airway epithelium. JCI Insight 2024; 9:e174888. [PMID: 38916962 PMCID: PMC11383597 DOI: 10.1172/jci.insight.174888] [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: 08/16/2023] [Accepted: 06/20/2024] [Indexed: 06/27/2024] Open
Abstract
The number of adults living with cystic fibrosis (CF) has already increased significantly because of drastic improvements in life expectancy attributable to advances in treatment, including the development of highly effective modulator therapy. Chronic airway inflammation in CF contributes to morbidity and mortality, and aging processes like inflammaging and cell senescence influence CF pathology. Our results show that single-cell RNA sequencing data, human primary bronchial epithelial cells from non-CF and CF donors, a CF bronchial epithelial cell line, and Cftr-knockout (Cftr-/-) rats all demonstrated increased cell senescence markers in the CF bronchial epithelium. This was associated with upregulation of fibroblast growth factor receptors (FGFRs) and mitogen-activated protein kinase (MAPK) p38. Inhibition of FGFRs, specifically FGFR4 and to some extent FGFR1, attenuated cell senescence and improved mucociliary clearance, which was associated with MAPK p38 signaling. Mucociliary dysfunction could also be improved using a combination of senolytics in a CF ex vivo model. In summary, FGFR/MAPK p38 signaling contributes to cell senescence in CF airways, which is associated with impaired mucociliary clearance. Therefore, attenuation of cell senescence in the CF airways might be a future therapeutic strategy improving mucociliary dysfunction and lung disease in an aging population with CF.
Collapse
Affiliation(s)
- Molly Easter
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine
| | - Meghan June Hirsch
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine
| | - Elex Harris
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine
- Gregory Fleming James Cystic Fibrosis Research Center, and
| | - Patrick Henry Howze
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine
| | - Emma Lea Matthews
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine
| | - Luke I. Jones
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine
| | - Seth Bollenbecker
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine
| | - Shia Vang
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine
| | - Daniel J. Tyrrell
- Division of Molecular and Cellular Pathology, Department of Pathology, The University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA
| | | | - Susan E. Birket
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine
- Gregory Fleming James Cystic Fibrosis Research Center, and
| | - Jarrod W. Barnes
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine
| | - Stefanie Krick
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine
- Gregory Fleming James Cystic Fibrosis Research Center, and
| |
Collapse
|
3
|
Bi Y, Zheng R, Hu J, Shi R, Shi J, Wang Y, Wang P, Jiang W, Kim G, Liu Z, Li X, Lin L. A novel FGFR1 inhibitor CYY292 suppresses tumor progression, invasion, and metastasis of glioblastoma by inhibiting the Akt/GSK3β/snail signaling axis. Genes Dis 2024; 11:479-494. [PMID: 37588207 PMCID: PMC10425802 DOI: 10.1016/j.gendis.2023.02.035] [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: 11/11/2022] [Accepted: 02/12/2023] [Indexed: 04/05/2023] Open
Abstract
Glioblastoma (GBM) is a malignant brain tumor that grows quickly, spreads widely, and is resistant to treatment. Fibroblast growth factor receptor (FGFR)1 is a receptor tyrosine kinase that regulates cellular processes, including proliferation, survival, migration, and differentiation. FGFR1 was predominantly expressed in GBM tissues, and FGFR1 expression was negatively correlated with overall survival. We rationally designed a novel small molecule CYY292, which exhibited a strong affinity for the FGFR1 protein in GBM cell lines in vitro. CYY292 also exerted an effect on the conserved Ser777 residue of FGFR1. CYY292 dose-dependently inhibited cell proliferation, epithelial-mesenchymal transition, stemness, invasion, and migration in vitro by specifically targeting the FGFR1/AKT/Snail pathways in GBM cells, and this effect was prevented by pharmacological inhibitors and critical gene knockdown. In vivo experiments revealed that CYY292 inhibited U87MG tumor growth more effectively than AZD4547. CYY292 also efficiently reduced GBM cell proliferation and increased survival in orthotopic GBM models. This study further elucidates the function of FGFR1 in the GBM and reveals the effect of CYY292, which targets FGFR1, on downstream signaling pathways directly reducing GBM cell growth, invasion, and metastasis and thus impairing the recruitment, activation, and function of immune cells.
Collapse
Affiliation(s)
- Yanran Bi
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
- College of Pharmacy and Research Institute of Drug Development, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Ruiling Zheng
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jiahao Hu
- Department of Dermatology, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou, Zhejiang 325000, China
| | - Ruiqing Shi
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Junfeng Shi
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yutao Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Peng Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Wenyi Jiang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Gyudong Kim
- College of Pharmacy and Research Institute of Drug Development, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Zhiguo Liu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiaokun Li
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Li Lin
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
- Zhejiang Lab for Regenerative Medicine, Oujiang Laboratory, Wenzhou, Zhejiang 325035, China
| |
Collapse
|
4
|
Woznowski MP, Potthoff SA, Königshausen E, Haase R, Hoch H, Meyer-Schwesinger C, Wiech T, Stegbauer J, Rump LC, Sellin L, Quack I. Inhibition of p38 MAPK decreases hyperglycemia-induced nephrin endocytosis and attenuates albuminuria. J Mol Med (Berl) 2022; 100:781-795. [PMID: 35451598 PMCID: PMC9110524 DOI: 10.1007/s00109-022-02184-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 01/20/2022] [Accepted: 02/22/2022] [Indexed: 11/26/2022]
Abstract
Abstract Chronic hyperglycemia, as in diabetes mellitus, may cause glomerular damage with microalbuminuria as an early sign. Noteworthy, even acute hyperglycemia can increase glomerular permeability before structural damage of the glomerular filter can be detected. Despite intensive research, specific antiproteinuric therapy is not available so far. Thus, a deeper understanding of the molecular mechanisms of albuminuria is desirable. P38 MAPK signaling is involved in the development of hyperglycemia-induced albuminuria. However, the mechanism of increased p38 MAPK activity leading to increased permeability and albuminuria remained unclear. Recently, we demonstrated that acute hyperglycemia triggers endocytosis of nephrin, the key molecule of the slit diaphragm, and induces albuminuria. Here, we identify p38 MAPK as a pivotal regulator of hyperglycemia-induced nephrin endocytosis. Activated p38 MAPK phosphorylates the nephrin c-terminus at serine 1146, facilitating the interaction of PKCα with nephrin. PKCα phosphorylates nephrin at threonine residues 1120 and 1125, mediating the binding of β-arrestin2 to nephrin. β-arrestin2 triggers endocytosis of nephrin by coupling it to the endocytic machinery, leading to increased glomerular permeability. Pharmacological inhibition of p38 MAPK preserves nephrin surface expression and significantly attenuates albuminuria. Key messages Acute hyperglycemia triggers endocytosis of nephrin. Activated p38 MAPK phosphorylates the nephrin c-terminus at serine 1146, facilitating the interaction of PKCα with nephrin. PKCα phosphorylates nephrin at threonine residues 1120 and 1125, mediating the binding of β-arrestin2 to nephrin. β-arrestin2 triggers endocytosis of nephrin by coupling it to the endocytic machinery, leading to a leaky glomerular filter. Pharmacological inhibition of p38 MAPK preserves nephrin surface expression and significantly attenuates albuminuria under hyperglycemic conditions.
Supplementary Information The online version contains supplementary material available at 10.1007/s00109-022-02184-5.
Collapse
Affiliation(s)
| | | | - Eva Königshausen
- Department of Nephrology, Medical Faculty, Heinrich-Heine University, 40225, Düsseldorf, Germany
| | - Raphael Haase
- Department of Nephrology, Medical Faculty, Heinrich-Heine University, 40225, Düsseldorf, Germany
| | - Henning Hoch
- Department of Nephrology, Medical Faculty, Heinrich-Heine University, 40225, Düsseldorf, Germany
| | - Catherine Meyer-Schwesinger
- Institute of Cellular and Integrative Physiology, University Clinic Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Thorsten Wiech
- Institute of Pathology, Nephropathology Section, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Johannes Stegbauer
- Department of Nephrology, Medical Faculty, Heinrich-Heine University, 40225, Düsseldorf, Germany
| | - Lars Christian Rump
- Department of Nephrology, Medical Faculty, Heinrich-Heine University, 40225, Düsseldorf, Germany
| | - Lorenz Sellin
- Department of Nephrology, Medical Faculty, Heinrich-Heine University, 40225, Düsseldorf, Germany
| | - Ivo Quack
- Emergency Department, Klinikum Konstanz, 78464, Konstanz, Germany
| |
Collapse
|
5
|
Mohale M, Gundampati RK, Krishnaswamy Suresh Kumar T, Heyes CD. Site-specific labeling and functional efficiencies of human fibroblast growth Factor-1 with a range of fluorescent Dyes in the flexible N-Terminal region and a rigid β-turn region. Anal Biochem 2022; 640:114524. [PMID: 34933004 DOI: 10.1016/j.ab.2021.114524] [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: 11/10/2021] [Accepted: 12/08/2021] [Indexed: 11/01/2022]
Abstract
Human fibroblast growth factor-1 (hFGF1) binding to its receptor and heparin play critical roles in cell proliferation, angiogenesis and wound healing but is also implicated in cancer. Fluorescence imaging is a powerful approach to study such protein interactions, but it is not always obvious if the site chosen will be efficiently labeled, often relying on trial-and-error. To provide a more systematic approach towards an efficient site-specific labeling strategy, we labeled two structurally distinct regions of the protein - the flexible N-terminus and a rigid loop. Several dyes were chosen to cover the visible region and to investigate how the structure of the dye affects the labeling efficiency. Flexibility in either the protein labeling site or the dye structure was found to result in high labeling efficiency, but flexibility in both resulted in a significant decrease in labeling efficiency. Conversely, too much rigidity in both can result in dye-protein interactions that can aggregate the protein. Importantly, site-specifically labeling hFGF1 in these regions maintained biological activity. These results could be applicable to other proteins by considering the flexibility of both the protein labeling site and the dye structure.
Collapse
Affiliation(s)
- Mamello Mohale
- Department of Chemistry and Biochemistry, University of Arkansas, 345 N. Campus Drive, Fayetteville, AR, 72701, USA
| | - Ravi Kumar Gundampati
- Department of Chemistry and Biochemistry, University of Arkansas, 345 N. Campus Drive, Fayetteville, AR, 72701, USA
| | | | - Colin D Heyes
- Department of Chemistry and Biochemistry, University of Arkansas, 345 N. Campus Drive, Fayetteville, AR, 72701, USA.
| |
Collapse
|
6
|
Hindle A, Bose C, Lee J, Palade PT, Peterson CJ, Reddy PH, Awasthi S, Singh SP. Rlip Depletion Alters Oncogene Transcription at Multiple Distinct Regulatory Levels. Cancers (Basel) 2022; 14:cancers14030527. [PMID: 35158795 PMCID: PMC8833773 DOI: 10.3390/cancers14030527] [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: 11/30/2021] [Revised: 01/07/2022] [Accepted: 01/15/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Rlip76 is a multifunctional membrane protein that facilitates cancer growth, and its depletion kills cancer cells. We recently found that Rlip depletion also results in broad changes to oncogene and tumor suppressor transcription. The present studies were designed to decipher the unknown downstream signaling pathways and transcriptional regulatory mechanisms driving the effect. Building on prior findings that Rlip depletion induces broad methylomic changes, we found using bioluminescence reporter assays that depletion of Rlip also exerts transcriptional control over several cancer genes through methylation-independent changes in transcription factor-mediated activation of their promoter regions and through additional as yet unidentified mechanisms. These findings have important implications for Rlip-targeted cancer therapy. Abstract Rlip76 (Rlip) is a multifunctional membrane protein that facilitates the high metabolic rates of cancer cells through the efflux of toxic metabolites and other functions. Rlip inhibition or depletion results in broad-spectrum anti-cancer effects in vitro and in vivo. Rlip depletion effectively suppresses malignancy and causes global reversion of characteristic CpG island methylomic and transcriptomic aberrations in the p53-null mouse model of spontaneous carcinogenesis through incompletely defined signaling and transcriptomic mechanisms. The methylome and transcriptome are normally regulated by the concerted actions of several mechanisms that include chromatin remodeling, promoter methylation, transcription factor interactions, and miRNAs. The present studies investigated the interaction of Rlip depletion or inhibition with the promoter methylation and transcription of selected cancer-related genes identified as being affected by Rlip depletion in our previous studies. We constructed novel promoter CpG island/luciferase reporter plasmids that respond only to CpG methylation and transcription factors. We found that Rlip depletion regulated expression by a transcription factor-based mechanism that functioned independently of promoter CpG methylation, lipid peroxidation, and p53 status.
Collapse
Affiliation(s)
- Ashly Hindle
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (A.H.); (C.B.); (J.L.); (C.J.P.); (P.H.R.)
| | - Chhanda Bose
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (A.H.); (C.B.); (J.L.); (C.J.P.); (P.H.R.)
| | - Jihyun Lee
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (A.H.); (C.B.); (J.L.); (C.J.P.); (P.H.R.)
- Division of Hematology & Oncology, Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Philip T. Palade
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Christopher J. Peterson
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (A.H.); (C.B.); (J.L.); (C.J.P.); (P.H.R.)
| | - P. Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (A.H.); (C.B.); (J.L.); (C.J.P.); (P.H.R.)
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Speech, Language and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Sanjay Awasthi
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (A.H.); (C.B.); (J.L.); (C.J.P.); (P.H.R.)
- Division of Hematology & Oncology, Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- UMC Cancer Center, UMC Health System, Lubbock, TX 79415, USA
- Correspondence: (S.A.); (S.P.S.); Tel.: +1-806-743-3543 (S.A.); +1-806-743-1540 (S.P.S.)
| | - Sharda P. Singh
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (A.H.); (C.B.); (J.L.); (C.J.P.); (P.H.R.)
- Division of Hematology & Oncology, Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Correspondence: (S.A.); (S.P.S.); Tel.: +1-806-743-3543 (S.A.); +1-806-743-1540 (S.P.S.)
| |
Collapse
|
7
|
The canonical FGF-FGFR signaling system at the molecular level. POSTEP HIG MED DOSW 2022. [DOI: 10.2478/ahem-2021-0024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Extracellular signaling molecules, among them the fibroblast growth factors (FGFs), enable cells to communicate with neighboring cells. Such signaling molecules that receive and transmit a signal require specific tyrosine kinase receptors located at the cell surface (fibroblast growth factor receptors, FGFRs). The binding of a signaling molecule to its specific receptor results in receptor dimerization and conformational changes in the cytoplasmic part of the receptor. The conformational changes lead to trans-autophosphorylation of the tyrosine kinase domains of the receptors and subsequently to induction of several downstream signaling pathways and expression of appropriate genes. The signaling pathways activated by FGFs control and coordinate cell behaviors such as cell division, migration, differentiation, and cell death. FGFs and their transmembrane receptors are widely distributed in different tissues and participate in fundamental processes during embryonic, fetal, and adult human life. The human FGF/FGFR family comprises 22 ligands and 4 high affinity receptors. In addition, FGFs bind to low affinity receptors, heparan sulfate proteoglycans at the cell surface. The availability of appropriate ligand/receptor pair, combined with the co-receptor, initiates signaling. Inappropriate FGF/FGFR signaling can cause skeletal disorders, primarily dwarfism, craniofacial malformation syndromes, mood disorders, metabolic disorders, and Kallman syndrome. In addition, aberrations in FGF/FGFR signaling have already been reported in several types of malignant diseases. Knowledge about the molecular mechanisms of FGF/FGFR activation and signaling is necessary to understand the basis of these diseases.
Collapse
|
8
|
Knaup I, Symmank J, Bastian A, Neuss S, Pufe T, Jacobs C, Wolf M. Impact of FGF1 on human periodontal ligament fibroblast growth, osteogenic differentiation and inflammatory reaction in vitro. J Orofac Orthop 2021; 83:42-55. [PMID: 34874457 DOI: 10.1007/s00056-021-00363-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 10/20/2021] [Indexed: 02/06/2023]
Abstract
PURPOSE To investigate in vitro the impact of fibroblast growth factor 1 (FGF1) in comparison to ascorbic acid (AscA) on human periodontal ligament fibroblast (HPdLF) growth, their osteogenic differentiation, and modulation of their inflammatory reaction to mechanical stress. METHODS The influence of different concentrations of FGF1 (12.5-200 ng/mL) on growth and proliferation of HPdLF cells was analyzed over 20 days by counting cell numbers and the percentage of Ki67-positive cells. Quantitative expression analysis of genes encoding the osteogenic markers alkaline phosphatase (ALPL), Runt-related transcription factor 2 (RUNX2), osteocalcin (OCN), and osteopontin (OSP), as well as the fibroblast markers vimentin (VIM) and fibroblast-specific protein 1 (FSP1), was performed after 2 and 20 days of cultivation. Metabolic activity was determined by MTT assay. For comparison with AscA, 50 ng/mL FGF1 was used for stimulation for 2 and 20 days. Cell number, percentage of Ki67-positive cells, and expression of osteoblast- and fibroblast-specific genes were examined. Alkaline phosphatase activity was visualized by NBT/BCIP and calcium deposits were stained with alizarin red. Cytokine (IL‑6, IL‑8, COX2/PGE2) expression and secretion were analyzed by qPCR and ELISA in 6 h mechanically compressed HPdLF cultured for 2 days with FGF1 or ascorbic acid. RESULTS Higher concentrations of FGF1 promoted cell proliferation upon short-term stimulation, whereas prolonged treatment induced the expression of osteogenic markers even with low concentrations. AscA promotes cell growth more markedly than FGF1 in short-term cultures, whereas FGF1 induced osteogenic cell fate more strongly in long-term culture. Both factors induced an increased inflammatory response of HPdLF to mechanical compression. CONCLUSION Our data suggest that FGF1 promotes an osteogenic phenotype of HPdLF and limits inflammatory response to mechanical forces compared to AscA.
Collapse
Affiliation(s)
- Isabel Knaup
- Department of Orthodontics, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany.
| | - Judit Symmank
- Department of Orthodontics, Jena University Hospital, Jena, Germany
| | - Asisa Bastian
- Department of Orthodontics, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Sabine Neuss
- Helmholtz Institute for Biomedical Engineering, BioInterface Group, RWTH Aachen University, Aachen, Germany
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Thomas Pufe
- Department of Anatomy and Cell Biology, RWTH Aachen University Hospital, Wendlingweg 2, 52074, Aachen, Germany
| | - Collin Jacobs
- Department of Orthodontics, Jena University Hospital, Jena, Germany
| | - Michael Wolf
- Department of Orthodontics, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany
| |
Collapse
|
9
|
p38-MAPK-mediated translation regulation during early blastocyst development is required for primitive endoderm differentiation in mice. Commun Biol 2021; 4:788. [PMID: 34172827 PMCID: PMC8233355 DOI: 10.1038/s42003-021-02290-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 06/02/2021] [Indexed: 02/07/2023] Open
Abstract
Successful specification of the two mouse blastocyst inner cell mass (ICM) lineages (the primitive endoderm (PrE) and epiblast) is a prerequisite for continued development and requires active fibroblast growth factor 4 (FGF4) signaling. Previously, we identified a role for p38 mitogen-activated protein kinases (p38-MAPKs) during PrE differentiation, but the underlying mechanisms have remained unresolved. Here, we report an early blastocyst window of p38-MAPK activity that is required to regulate ribosome-related gene expression, rRNA precursor processing, polysome formation and protein translation. We show that p38-MAPK inhibition-induced PrE phenotypes can be partially rescued by activating the translational regulator mTOR. However, similar PrE phenotypes associated with extracellular signal-regulated kinase (ERK) pathway inhibition targeting active FGF4 signaling are not affected by mTOR activation. These data indicate a specific role for p38-MAPKs in providing a permissive translational environment during mouse blastocyst PrE differentiation that is distinct from classically reported FGF4-based mechanisms.
Collapse
|
10
|
Margiotta A. All Good Things Must End: Termination of Receptor Tyrosine Kinase Signal. Int J Mol Sci 2021; 22:ijms22126342. [PMID: 34198477 PMCID: PMC8231876 DOI: 10.3390/ijms22126342] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 12/28/2022] Open
Abstract
Receptor tyrosine kinases (RTKs) are membrane receptors that regulate many fundamental cellular processes. A tight regulation of RTK signaling is fundamental for development and survival, and an altered signaling by RTKs can cause cancer. RTKs are localized at the plasma membrane (PM) and the major regulatory mechanism of signaling of RTKs is their endocytosis and degradation. In fact, RTKs at the cell surface bind ligands with their extracellular domain, become active, and are rapidly internalized where the temporal extent of signaling, attenuation, and downregulation are modulated. However, other mechanisms of signal attenuation and termination are known. Indeed, inhibition of RTKs’ activity may occur through the modulation of the phosphorylation state of RTKs and the interaction with specific proteins, whereas antagonist ligands can inhibit the biological responses mediated by the receptor. Another mechanism concerns the expression of endogenous inactive receptor variants that are deficient in RTK activity and take part to inactive heterodimers or hetero-oligomers. The downregulation of RTK signals is fundamental for several cellular functions and the homeostasis of the cell. Here, we will review the mechanisms of signal attenuation and termination of RTKs, focusing on FGFRs.
Collapse
Affiliation(s)
- Azzurra Margiotta
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic;
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czech Republic
| |
Collapse
|
11
|
Mao L, Liao C, Qin J, Gong Y, Zhou Y, Li S, Liu Z, Deng H, Deng W, Sun Q, Mo X, Xue Y, Billadeau DD, Dai L, Li G, Jia D. Phosphorylation of SNX27 by MAPK11/14 links cellular stress-signaling pathways with endocytic recycling. J Cell Biol 2021; 220:211812. [PMID: 33605979 PMCID: PMC7901142 DOI: 10.1083/jcb.202010048] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/28/2020] [Accepted: 01/21/2021] [Indexed: 02/08/2023] Open
Abstract
Endocytosed proteins can be delivered to lysosomes for degradation or recycled to either the trans-Golgi network or the plasma membrane. It remains poorly understood how the recycling versus degradation of cargoes is determined. Here, we show that multiple extracellular stimuli, including starvation, LPS, IL-6, and EGF treatment, can strongly inhibit endocytic recycling of multiple cargoes through the activation of MAPK11/14. The stress-induced kinases in turn directly phosphorylate SNX27, a key regulator of endocytic recycling, at serine 51 (Ser51). Phosphorylation of SNX27 at Ser51 alters the conformation of its cargo-binding pocket and decreases the interaction between SNX27 and cargo proteins, thereby inhibiting endocytic recycling. Our study indicates that endocytic recycling is highly dynamic and can crosstalk with cellular stress–signaling pathways. Suppression of endocytic recycling and enhancement of receptor lysosomal degradation serve as new mechanisms for cells to cope with stress and save energy.
Collapse
Affiliation(s)
- Lejiao Mao
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Chenyi Liao
- Laboratory of Molecular Modeling and Design, State Key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Jiao Qin
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Yanqiu Gong
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yifei Zhou
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Shasha Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Zhe Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Huaqing Deng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Wankun Deng
- Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Qingxiang Sun
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xianming Mo
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Xue
- Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Daniel D Billadeau
- Division of Oncology Research and Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN
| | - Lunzhi Dai
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Guohui Li
- Laboratory of Molecular Modeling and Design, State Key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Da Jia
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| |
Collapse
|
12
|
Disabled-2: a positive regulator of the early differentiation of myoblasts. Cell Tissue Res 2020; 381:493-508. [PMID: 32607799 PMCID: PMC7431403 DOI: 10.1007/s00441-020-03237-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 06/05/2020] [Indexed: 11/25/2022]
Abstract
Dab2 is an adaptor protein and a tumor suppressor. Our previous study has found that Dab2 was expressed in early differentiating skeletal muscles in mouse embryos. In this study, we determined the role of Dab2 in the skeletal muscle differentiation using C2C12 myoblasts in vitro and Xenopus laevis embryos in vivo. The expression of Dab2 was increased in C2C12 myoblasts during the formation of myotubes in vitro. Knockdown of Dab2 expression in C2C12 myoblasts resulted in a reduction of myotube formation, whereas the myotube formation was enhanced upon overexpression of Dab2. Re-expression of Dab2 in C2C12 myoblasts with downregulated expression of Dab2 restored their capacity to form myotubes. Microarray profiling and subsequent network analyses on the 155 differentially expressed genes after Dab2 knockdown showed that Mef2c was an important myogenic transcription factor regulated by Dab2 through the p38 MAPK pathway. It was also involved in other pathways that are associated with muscular development and functions. In Xenopus embryos developed in vivo, XDab2 was expressed in the myotome of somites where various myogenic markers were also expressed. Knockdown of XDab2 expression with antisense morpholinos downregulated the expression of myogenic markers in somites. In conclusion, this study is the first to provide solid evidence to show that Dab2 is a positive regulator of the early myoblast differentiation.
Collapse
|
13
|
Han J, Wu J, Silke J. An overview of mammalian p38 mitogen-activated protein kinases, central regulators of cell stress and receptor signaling. F1000Res 2020; 9. [PMID: 32612808 PMCID: PMC7324945 DOI: 10.12688/f1000research.22092.1] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/18/2020] [Indexed: 12/19/2022] Open
Abstract
The p38 family is a highly evolutionarily conserved group of mitogen-activated protein kinases (MAPKs) that is involved in and helps co-ordinate cellular responses to nearly all stressful stimuli. This review provides a succinct summary of multiple aspects of the biology, role, and substrates of the mammalian family of p38 kinases. Since p38 activity is implicated in inflammatory and other diseases, we also discuss the clinical implications and pharmaceutical approaches to inhibit p38.
Collapse
Affiliation(s)
- Jiahuai Han
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China
| | - Jianfeng Wu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China
| | - John Silke
- The Walter and Eliza Hall Institute, IG Royal Parade, Parkville, Victoria, 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, 3050, Australia
| |
Collapse
|
14
|
Sluzalska KD, Slawski J, Sochacka M, Lampart A, Otlewski J, Zakrzewska M. Intracellular partners of fibroblast growth factors 1 and 2 - implications for functions. Cytokine Growth Factor Rev 2020; 57:93-111. [PMID: 32475760 DOI: 10.1016/j.cytogfr.2020.05.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/04/2020] [Accepted: 05/07/2020] [Indexed: 01/01/2023]
Abstract
Fibroblast growth factors 1 and 2 (FGF1 and FGF2) are mainly considered as ligands of surface receptors through which they regulate a broad spectrum of biological processes. They are secreted in non-canonical way and, unlike other growth factors, they are able to translocate from the endosome to the cell interior. These unique features, as well as the role of the intracellular pool of FGF1 and FGF2, are far from being fully understood. An increasing number of reports address this problem, focusing on the intracellular interactions of FGF1 and 2. Here, we summarize the current state of knowledge of the FGF1 and FGF2 binding partners inside the cell and the possible role of these interactions. The partner proteins are grouped according to their function, including proteins involved in secretion, cell signaling, nucleocytoplasmic transport, binding and processing of nucleic acids, ATP binding, and cytoskeleton assembly. An in-depth analysis of the network of these binding partners could indicate novel, non-classical functions of FGF1 and FGF2 and uncover an additional level of a fine control of the well-known FGF-regulated cellular processes.
Collapse
Affiliation(s)
- Katarzyna Dominika Sluzalska
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Jakub Slawski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Martyna Sochacka
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Agata Lampart
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Jacek Otlewski
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Malgorzata Zakrzewska
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| |
Collapse
|
15
|
Analysis of potential roles of combinatorial microRNA regulation in occurrence of valvular heart disease with atrial fibrillation based on computational evidences. PLoS One 2019; 14:e0221900. [PMID: 31479479 PMCID: PMC6719876 DOI: 10.1371/journal.pone.0221900] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 08/16/2019] [Indexed: 11/19/2022] Open
Abstract
Background Atrial fibrillation (AF) is the most common arrhythmia. Patients with valvular heart disease (VHD) frequently have AF. Growing evidence demonstrates that a specifically altered pattern of microRNA (miRNA) expression is related to valvular heart disease with atrial fibrillation (AF-VHD) processes. However, the combinatorial regulation by multiple miRNAs in inducing AF-VHD remains largely unknown. Methods The work identified AF-VHD-specific miRNAs and their combinations through mapping miRNA expression profile into differential co-expression network. The expressions of some dysregulated miRNAs were measured by quantitative reverse transcription polymerase chain reaction (qRT-PCR). The regulations of signaling pathways by the combinatorial miRNAs were predicted by enrichment analysis tools. Results Thirty-two differentially expressed (DE) miRNAs were identified to be AF-VHD-specific, some of which were new findings. These miRNAs interacted to form 5 combinations. qRT-PCR confirmed the different expression of several identified miRNAs, which illustrated the reliability and biomarker potentials of 32 dysregulation miRNAs. The biological characteristics of combinatorial miRNAs related to AF-VHD were highlighted. Twelve signaling pathways regulated by combinatorial miRNAs were predicted to be possibly associated with AF-VHD. Conclusions The AF-VHD-related signaling pathways regulated by combinatorial miRNAs may play an important role in the occurrence of AF-VHD. The work brings new insights into biomarkers and miRNA combination regulation mechanism in AF-VHD as well as further biological experiments.
Collapse
|
16
|
Zarredar H, Farajnia S, Ansarin K, Baradaran B, Aria M, Asadi M. Synergistic Effect of Novel EGFR Inhibitor AZD8931 and p38α siRNA in Lung Adenocarcinoma Cancer Cells. Anticancer Agents Med Chem 2019; 19:638-644. [DOI: 10.2174/1871520619666190301125203] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/26/2018] [Accepted: 02/13/2019] [Indexed: 12/19/2022]
Abstract
Background:Lung cancer is the leading cause of cancer-related death with less than 5-year survival rate for both men and women worldwide. EGFR and MAPK signaling pathways have a critical role in proliferation and progression of various cancers, including lung cancer. P38 map kinase plays different role in various tissue hence showing a tissue-dependent behavior. It acts as an oncogene in some tissues while plays as tumor suppressor in some other tissues. The aim of this study was to investigate the combined effect of P38 αspecific siRNA and EGFR inhibitor on apoptosis and proliferation of A549 lung cancer cell line.Objective:This article is dedicated to the synergistic effect of novel EGFR inhibitor AZD8931 and P38 α siRNA in lung adenocarcinoma cancer cells proliferation and apoptosis.Methods and Materials:The A549 lung cancer cells were treated with P38 α- siRNA and EGFR inhibitor alone or in combination. The cytotoxic effects of P38 α- siRNA and EGFR inhibitor were determined using MTT assay. Relative P38 α and EGFR mRNA levels were measured by QRT-PCR. Induction of apoptosis were measured by FACS analysis.Results:The expression of mRNA related to P38 α, EGFR, and Her2 genes was reduced to 23.4%, 52.4%, and 75, respectively, after treatment of their inhibitors. Also, MTT assay showed that the cell viability after treatment with p38 α SiRNA, EGFR inhibitor and their combination was reduced to 51.02%, 48.9%, and 25.11%, respectively. FACS results indicated that p38 α siRNA, EGFR inhibitor and their combination, reduced the population of live cells to 49.5%, 32.2% and 14.3% in comparison to the population of untreated control cells (99.5%).Conclusion:The results of this study indicated that p38 α and EGFR might play an important role in the development and growth of lung cancer and might be a potential therapeutic target for the treatment of lung cancer.
Collapse
Affiliation(s)
- Habib Zarredar
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Science. Tabriz, Iran
| | - Safar Farajnia
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Science. Tabriz, Iran
| | - Khalil Ansarin
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Science. Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Aria
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milad Asadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
17
|
Survey of cellular immune responses to human cytomegalovirus infection in the microenvironment of the uterine-placental interface. Med Microbiol Immunol 2019; 208:475-485. [PMID: 31065796 DOI: 10.1007/s00430-019-00613-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 04/09/2019] [Indexed: 12/19/2022]
Abstract
Congenital human cytomegalovirus (HCMV) infection is a leading cause of birth defects, yet there are no established treatments for preventing maternal-fetal transmission. During first trimester, HCMV replicates in basal decidua that functions as a reservoir for virus and source of transmission to the attached placenta and fetal hemiallograft but also contains immune cells, including natural killer cells, macrophages, and T cell subsets, that respond to pathogens, protecting the placenta and fetus. However, the specific cellular and cytokine responses to infection are unknown, nor are the immune correlates of protection that guide development of therapeutic strategies. Here we survey immune cell phenotypes in intact explants of basal decidua infected with a clinical pathogenic HCMV strain ex vivo and identify specific changes occurring in response to infection in the tissue environment. Using 4-color immunofluorescence microscopy, we found that at 3 days postinfection, virus replicates in decidual stromal cells and epithelial cells of endometrial glands. Infected cells and effector memory CD8+ T cells (TEM) in contact with them make IFN-γ. CD8+ TEM cells produce granulysin and cluster at sites of infection in decidua and the epithelium of endometrial glands. Quantification indicated expansion of two immune cell subtypes-CD8+ TEM cells and, to a lesser extent, iNKT cells. Approximately 20% of immune cells were found in pairs in both control and infected decidua, suggesting frequent cross-talk in the microenvironment of decidua. Our findings indicate a complex immune microenvironment in basal decidua and suggest CD8+ TEM cells play a role in early responses to decidual infection in seropositive women.
Collapse
|
18
|
Crosstalk between p38 and Erk 1/2 in Downregulation of FGF1-Induced Signaling. Int J Mol Sci 2019; 20:ijms20081826. [PMID: 31013829 PMCID: PMC6514807 DOI: 10.3390/ijms20081826] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 01/25/2019] [Accepted: 04/10/2019] [Indexed: 11/16/2022] Open
Abstract
Mitogen-activated protein kinases (MAPK): Erk1 and Erk2 are key players in negative-feedback regulation of fibroblast growth factor (FGF) signaling. Upon activation, Erk1 and Erk2 directly phosphorylate FGF receptor 1 (FGFR1) at a specific serine residue in the C-terminal part of the receptor, substantially reducing the tyrosine phosphorylation in the receptor kinase domain and its signaling. Similarly, active Erks can also phosphorylate multiple threonine residues in the docking protein FGF receptor substrate 2 (FRS2), a major mediator of FGFR signaling. Here, we demonstrate that in NIH3T3 mouse fibroblasts and human osteosarcoma U2OS cells stably expressing FGFR1, in addition to Erk1 and Erk2, p38 kinase is able to phosphorylate FRS2. Simultaneous inhibition of Erk1/2 and p38 kinase led to a significant change in the phosphorylation pattern of FRS2 that in turn resulted in prolonged tyrosine phosphorylation of FGFR1 and FRS2 and in sustained signaling, as compared to the selective inhibition of Erks. Furthermore, excessive activation of p38 with anisomycin partially compensated the lack of Erks activity. These experiments reveal a novel crosstalk between p38 and Erk1/2 in downregulation of FGF-induced signaling.
Collapse
|
19
|
Zarredar H, Pashapour S, Farajnia S, Ansarin K, Baradaran B, Ahmadzadeh V, Safari F. Targeting the KRAS, p38α, and NF-κB in lung adenocarcinoma cancer cells: The effect of combining RNA interferences with a chemical inhibitor. J Cell Biochem 2019; 120:10670-10677. [PMID: 30656741 DOI: 10.1002/jcb.28357] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 11/29/2018] [Indexed: 12/28/2022]
Abstract
BACKGROUND Lung cancer is the leading cause of cancer-related death with less than 5-year survival rate for both men and women worldwide. KRAS (Kirsten rat sarcoma), nuclear factor-κB (NF-κB), and mitogen-activated protein kinase (MAPK) signaling pathways have a critical role in the proliferation and progression of various cancers, including lung cancer. The p38 MAPK plays a different role in various tissue hence show a tissue-dependent behavior. It acts as an oncogene in some tissues while plays as a tumor suppressor in some other tissues. Also, KRAS and NF-κB act as an oncogene in various cancer. This study was dedicated to analyzing the combined effect of NF-κB inhibitor, specific KRAS, and p38α small interfering RNA (siRNA) in A549 cell line. MATERIALS AND METHODS The cytotoxic effects of p38α siRNA, KRAS siRNA, and NF-κB inhibitor were determined using the 3-(4,5-dimethylthiazol-2-yl)-2,5 diphenyl tetrazolium bromide (MTT) assay. Relative p38α, KRAS, and NF-κB messenger RNA (mRNA) levels were measured by quantitative reverse-transcription polymerase chain reaction. Induction of apoptosis by treatments was measured by fluorescence-activated cell sorting (FACS) analysis. RESULTS The expression of mRNA related to p38α and KRAS genes was reduced to 23.4% and 26.7%, respectively, after treatment with specific siRNAs. Also, MTT assay showed that the cell viability after treatment with p38α siRNA, KRAS siRNA, NF-κB inhibitor and their combination was reduced. FACS results indicated that p38α siRNA, KRAS siRNA, and NF-κB inhibitor, and their combination, reduced the population of live cells in comparison with the population of untreated control cells (99.5%). The results are expressed as mean ± SD (n = 3); *P < 0.05; ** P < 0.01; *** P < 0.001; **** P < 0.0001 vs control group. CONCLUSION The results of this study indicated that p38α, KRAS, and NF-κB signaling pathways might play an important role in the development and growth of lung cancer and might be a potential therapeutic target for treatment of lung cancer.
Collapse
Affiliation(s)
- Habib Zarredar
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Science, Tabriz, Iran.,Students Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shadi Pashapour
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Science, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Safar Farajnia
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Science, Tabriz, Iran.,Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khalil Ansarin
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahideh Ahmadzadeh
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Safari
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
20
|
Dinsmore CJ, Soriano P. MAPK and PI3K signaling: At the crossroads of neural crest development. Dev Biol 2018; 444 Suppl 1:S79-S97. [PMID: 29453943 PMCID: PMC6092260 DOI: 10.1016/j.ydbio.2018.02.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 02/06/2018] [Accepted: 02/06/2018] [Indexed: 02/08/2023]
Abstract
Receptor tyrosine kinase-mediated growth factor signaling is essential for proper formation and development of the neural crest. The many ligands and receptors implicated in these processes signal through relatively few downstream pathways, frequently converging on the MAPK and PI3K pathways. Despite decades of study, there is still considerable uncertainty about where and when these signaling pathways are required and how they elicit particular responses. This review summarizes our current understanding of growth factor-induced MAPK and PI3K signaling in the neural crest.
Collapse
Affiliation(s)
- Colin J Dinsmore
- Department of Cell, Developmental and Regenerative Biology, Tisch Cancer Institute, Icahn School of Medicine at Mt. Sinai, New York, NY 10029, USA
| | - Philippe Soriano
- Department of Cell, Developmental and Regenerative Biology, Tisch Cancer Institute, Icahn School of Medicine at Mt. Sinai, New York, NY 10029, USA.
| |
Collapse
|
21
|
Kostas M, Lampart A, Bober J, Wiedlocha A, Tomala J, Krowarsch D, Otlewski J, Zakrzewska M. Translocation of Exogenous FGF1 and FGF2 Protects the Cell against Apoptosis Independently of Receptor Activation. J Mol Biol 2018; 430:4087-4101. [DOI: 10.1016/j.jmb.2018.08.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 07/30/2018] [Accepted: 08/06/2018] [Indexed: 01/16/2023]
|
22
|
Biomaterials in Tendon and Skeletal Muscle Tissue Engineering: Current Trends and Challenges. MATERIALS 2018; 11:ma11071116. [PMID: 29966303 PMCID: PMC6073924 DOI: 10.3390/ma11071116] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 06/20/2018] [Accepted: 06/25/2018] [Indexed: 12/17/2022]
Abstract
Tissue engineering is a promising approach to repair tendon and muscle when natural healing fails. Biohybrid constructs obtained after cells’ seeding and culture in dedicated scaffolds have indeed been considered as relevant tools for mimicking native tissue, leading to a better integration in vivo. They can also be employed to perform advanced in vitro studies to model the cell differentiation or regeneration processes. In this review, we report and analyze the different solutions proposed in literature, for the reconstruction of tendon, muscle, and the myotendinous junction. They classically rely on the three pillars of tissue engineering, i.e., cells, biomaterials and environment (both chemical and physical stimuli). We have chosen to present biomimetic or bioinspired strategies based on understanding of the native tissue structure/functions/properties of the tissue of interest. For each tissue, we sorted the relevant publications according to an increasing degree of complexity in the materials’ shape or manufacture. We present their biological and mechanical performances, observed in vitro and in vivo when available. Although there is no consensus for a gold standard technique to reconstruct these musculo-skeletal tissues, the reader can find different ways to progress in the field and to understand the recent history in the choice of materials, from collagen to polymer-based matrices.
Collapse
|
23
|
Thamodaran V, Bruce AW. p38 (Mapk14/11) occupies a regulatory node governing entry into primitive endoderm differentiation during preimplantation mouse embryo development. Open Biol 2017; 6:rsob.160190. [PMID: 27605380 PMCID: PMC5043583 DOI: 10.1098/rsob.160190] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 08/12/2016] [Indexed: 12/31/2022] Open
Abstract
During mouse preimplantation embryo development, the classically described second cell-fate decision involves the specification and segregation, in blastocyst inner cell mass (ICM), of primitive endoderm (PrE) from pluripotent epiblast (EPI). The active role of fibroblast growth factor (Fgf) signalling during PrE differentiation, particularly in the context of Erk1/2 pathway activation, is well described. However, we report that p38 family mitogen-activated protein kinases (namely p38α/Mapk14 and p38β/Mapk11; referred to as p38-Mapk14/11) also participate in PrE formation. Specifically, functional p38-Mapk14/11 are required, during early-blastocyst maturation, to assist uncommitted ICM cells, expressing both EPI and earlier PrE markers, to fully commit to PrE differentiation. Moreover, functional activation of p38-Mapk14/11 is, as reported for Erk1/2, under the control of Fgf-receptor signalling, plus active Tak1 kinase (involved in non-canonical bone morphogenetic protein (Bmp)-receptor-mediated PrE differentiation). However, we demonstrate that the critical window of p38-Mapk14/11 activation precedes the E3.75 timepoint (defined by the initiation of the classical ‘salt and pepper’ expression pattern of mutually exclusive EPI and PrE markers), whereas appropriate lineage maturation is still achievable when Erk1/2 activity (via Mek1/2 inhibition) is limited to a period after E3.75. We propose that active p38-Mapk14/11 act as enablers, and Erk1/2 as drivers, of PrE differentiation during ICM lineage specification and segregation.
Collapse
Affiliation(s)
- Vasanth Thamodaran
- Laboratory of Developmental Biology and Genetics (LDB&G), Department of Molecular Biology, Faculty of Science, University of South Bohemia, Branišovská 31, 37005 České Budějovice, Czech Republic
| | - Alexander W Bruce
- Laboratory of Developmental Biology and Genetics (LDB&G), Department of Molecular Biology, Faculty of Science, University of South Bohemia, Branišovská 31, 37005 České Budějovice, Czech Republic Biology Centre of the Czech Academy of Sciences, Institute of Entomology, Branišovská 31, 37005 České Budějovice, Czech Republic
| |
Collapse
|
24
|
MuSK Kinase Activity is Modulated By A Serine Phosphorylation Site in The Kinase Loop. Sci Rep 2016; 6:33583. [PMID: 27666825 PMCID: PMC5035991 DOI: 10.1038/srep33583] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 08/31/2016] [Indexed: 11/16/2022] Open
Abstract
The neuromuscular junction (NMJ) forms when a motor neuron contacts a muscle fibre. A reciprocal exchange of signals initiates a cascade of signalling events that result in pre- and postsynaptic differentiation. At the centre of these signalling events stands muscle specific kinase (MuSK). MuSK activation, kinase activity and subsequent downstream signalling are crucial for NMJ formation as well as maintenance. Therefore MuSK kinase activity is tightly regulated to ensure proper NMJ development. We have identified a novel serine phosphorylation site at position 751 in MuSK that is increasingly phosphorylated upon agrin stimulation. S751 is also phosphorylated in muscle tissue and its phosphorylation depends on MuSK kinase activity. A phosphomimetic mutant of S751 increases MuSK kinase activity in response to non-saturating agrin concentrations . In addition, basal MuSK and AChR phosphorylation as well as AChR cluster size are increased. We believe that the phosphorylation of S751 provides a novel mechanism to relief the autoinhibition of the MuSK activation loop. Such a lower autoinhibition could foster or stabilize MuSK kinase activation, especially during stages when no or low level of agrin are present. Phosphorylation of S751 might therefore represent a novel mechanism to modulate MuSK kinase activity during prepatterning or NMJ maintenance.
Collapse
|
25
|
Abstract
AXL is a tyrosine kinase receptor activated by GAS6 and regulates cancer cell proliferation migration and angiogenesis. We studied AXL as new therapeutic target in colorectal cancer (CRC). Expression and activation of AXL and GAS6 were evaluated in a panel of human CRC cell lines. AXL gene silencing or pharmacologic inhibition with foretinib suppressed proliferation, migration and survival in CRC cells. In an orthotopic colon model of human HCT116 CRC cells overexpressing AXL, foretinib treatment caused significant inhibition of tumour growth and peritoneal metastatic spreading. AXL and GAS6 overexpression by immunohistochemistry (IHC) were found in 76,7% and 73.5%, respectively, of 223 human CRC specimens, correlating with less differentiated histological grading. GAS6 overexpression was associated with nodes involvement and tumour stage. AXL gene was found amplified by Fluorescence in situ hybridization (FISH) in 8/146 cases (5,4%) of CRC samples. Taken together, AXL inhibition could represent a novel therapeutic approach in CRC.
Collapse
|
26
|
Qaddoumi I, Orisme W, Wen J, Santiago T, Gupta K, Dalton JD, Tang B, Haupfear K, Punchihewa C, Easton J, Mulder H, Boggs K, Shao Y, Rusch M, Becksfort J, Gupta P, Wang S, Lee RP, Brat D, Peter Collins V, Dahiya S, George D, Konomos W, Kurian KM, McFadden K, Serafini LN, Nickols H, Perry A, Shurtleff S, Gajjar A, Boop FA, Klimo PD, Mardis ER, Wilson RK, Baker SJ, Zhang J, Wu G, Downing JR, Tatevossian RG, Ellison DW. Genetic alterations in uncommon low-grade neuroepithelial tumors: BRAF, FGFR1, and MYB mutations occur at high frequency and align with morphology. Acta Neuropathol 2016; 131:833-45. [PMID: 26810070 DOI: 10.1007/s00401-016-1539-z] [Citation(s) in RCA: 262] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Revised: 01/16/2016] [Accepted: 01/17/2016] [Indexed: 12/24/2022]
Abstract
Low-grade neuroepithelial tumors (LGNTs) are diverse CNS tumors presenting in children and young adults, often with a history of epilepsy. While the genetic profiles of common LGNTs, such as the pilocytic astrocytoma and 'adult-type' diffuse gliomas, are largely established, those of uncommon LGNTs remain to be defined. In this study, we have used massively parallel sequencing and various targeted molecular genetic approaches to study alterations in 91 LGNTs, mostly from children but including young adult patients. These tumors comprise dysembryoplastic neuroepithelial tumors (DNETs; n = 22), diffuse oligodendroglial tumors (d-OTs; n = 20), diffuse astrocytomas (DAs; n = 17), angiocentric gliomas (n = 15), and gangliogliomas (n = 17). Most LGNTs (84 %) analyzed by whole-genome sequencing (WGS) were characterized by a single driver genetic alteration. Alterations of FGFR1 occurred frequently in LGNTs composed of oligodendrocyte-like cells, being present in 82 % of DNETs and 40 % of d-OTs. In contrast, a MYB-QKI fusion characterized almost all angiocentric gliomas (87 %), and MYB fusion genes were the most common genetic alteration in DAs (41 %). A BRAF:p.V600E mutation was present in 35 % of gangliogliomas and 18 % of DAs. Pathogenic alterations in FGFR1/2/3, BRAF, or MYB/MYBL1 occurred in 78 % of the series. Adult-type d-OTs with an IDH1/2 mutation occurred in four adolescents, the youngest aged 15 years at biopsy. Despite a detailed analysis, novel genetic alterations were limited to two fusion genes, EWSR1-PATZ1 and SLMAP-NTRK2, both in gangliogliomas. Alterations in BRAF, FGFR1, or MYB account for most pathogenic alterations in LGNTs, including pilocytic astrocytomas, and alignment of these genetic alterations and cytologic features across LGNTs has diagnostic implications. Additionally, therapeutic options based upon targeting the effects of these alterations are already in clinical trials.
Collapse
|
27
|
Bober J, Olsnes S, Kostas M, Bogacz M, Zakrzewska M, Otlewski J. Identification of new FGF1 binding partners-Implications for its intracellular function. IUBMB Life 2016; 68:242-51. [PMID: 26840910 PMCID: PMC4832500 DOI: 10.1002/iub.1480] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 01/07/2016] [Indexed: 11/30/2022]
Abstract
Besides its classical mode of action through activation of specific receptors at the cell surface, fibroblast growth factor 1 (FGF1) can also cross the cellular membrane and translocate into the cytosol and further to the nucleus. The mechanism of this translocation is described partially, but the role of FGF1 inside the cell remains unknown. The aim of our work was to identify novel binding partners of FGF1 to predict its intracellular functions. We combined three methods of identification of such partners based on different principles: yeast two‐hybrid screen and mass spectrometry (MS) analysis of complexes obtained by Tandem Affinity Purification (TAP) or by co‐precipitation from cell lysate using recombinant FGF1. Altogether, we identified twenty novel intracellular proteins interacting with FGF1. For selected proteins, their direct interaction with FGF1 was confirmed by pull‐down assays and SPR measurements. Interestingly, half of the proteins found are involved in processes related to cell viability, such as apoptosis, cell proliferation, and cell cycle regulation. Thus, our study indicates that the role of intracellular FGF1 is to protect the cell against stress conditions by providing an additional signal for cell survival, independently of receptor‐activated signaling cascades. © 2016 IUBMB Life, 68(3):242–251, 2016
Collapse
Affiliation(s)
- Joanna Bober
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Wroclaw, Poland
| | - Sjur Olsnes
- Department of Biochemistry, Institute for Cancer Research, Oslo University Hospital, Montebello, Oslo, Norway
| | - Michal Kostas
- Faculty of Biotechnology, Department of Protein Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Marek Bogacz
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Wroclaw, Poland
| | - Malgorzata Zakrzewska
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Wroclaw, Poland
| | - Jacek Otlewski
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Wroclaw, Poland
| |
Collapse
|
28
|
Chou SM, Lai WJ, Hong T, Tsai SH, Chen YH, Kao CH, Chu R, Shen TL, Li TK. Involvement of p38 MAPK in the Anticancer Activity of Cultivated Cordyceps militaris. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2015. [PMID: 26205966 DOI: 10.1142/s0192415x15500603] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Cordyceps militaris is a traditional Chinese medicine frequently used for tonic and therapeutic purposes. Reports from our laboratory and others have demonstrated that extracts of the cultivated fruiting bodies of C. militaris (CM) exhibit a potent cytotoxic effect against many cancer cell lines, especially human leukemia cells. Here, we further investigated the underlying mechanism through which CM is cytotoxic to cancer cells. The CM-mediated induction of PARP cleavage and its related DNA damage signal (γH2AX) was diminished by caspase inhibitor I. In contrast, a ROS scavenger failed to prevent CM-mediated leukemia cell death. Moreover, two signaling molecules, AKT and p38 MAPK, were activated during the course of apoptosis induction. Employing MTT analysis, we found that a p38 MAPK inhibitor but not an AKT inhibitor could rescue cells from CM-mediated cell death, as well as inhibit the cleavage of PARP, formation of apoptotic bodies and up-regulation of the γH2AX signal. These results suggest that CM-mediated leukemia cell death occurs through the activation of the p38 MAPK pathway, indicating its potential therapeutic effects against human leukemia.
Collapse
Affiliation(s)
- Shang-Min Chou
- Department and Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | | | - Tzuwen Hong
- Mucho Biotechnology Inc., Taipei 10684, Taiwan.,Department of Plant Pathology and Microbiology, National Taiwan University, Taipei 10617, Taiwan
| | | | | | | | - Richard Chu
- Mucho Biotechnology Inc., Taipei 10684, Taiwan
| | - Tang-Long Shen
- Department of Plant Pathology and Microbiology, National Taiwan University, Taipei 10617, Taiwan.,Center for Biotechnology, National Taiwan University, Taipei 10617, Taiwan
| | - Tsai-Kun Li
- Department and Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan.,Center for Biotechnology, National Taiwan University, Taipei 10617, Taiwan
| |
Collapse
|
29
|
Sletten T, Kostas M, Bober J, Sorensen V, Yadollahi M, Olsnes S, Tomala J, Otlewski J, Zakrzewska M, Wiedlocha A. Nucleolin regulates phosphorylation and nuclear export of fibroblast growth factor 1 (FGF1). PLoS One 2014; 9:e90687. [PMID: 24595027 PMCID: PMC3942467 DOI: 10.1371/journal.pone.0090687] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 02/04/2014] [Indexed: 11/19/2022] Open
Abstract
Extracellular fibroblast growth factor 1 (FGF1) acts through cell surface tyrosine kinase receptors, but FGF1 can also act directly in the cell nucleus, as a result of nuclear import of endogenously produced, non-secreted FGF1 or by transport of extracellular FGF1 via endosomes and cytosol into the nucleus. In the nucleus, FGF1 can be phosphorylated by protein kinase C δ (PKCδ), and this event induces nuclear export of FGF1. To identify intracellular targets of FGF1 we performed affinity pull-down assays and identified nucleolin, a nuclear multifunctional protein, as an interaction partner of FGF1. We confirmed a direct nucleolin-FGF1 interaction by surface plasmon resonance and identified residues of FGF1 involved in the binding to be located within the heparin binding site. To assess the biological role of the nucleolin-FGF1 interaction, we studied the intracellular trafficking of FGF1. In nucleolin depleted cells, exogenous FGF1 was endocytosed and translocated to the cytosol and nucleus, but FGF1 was not phosphorylated by PKCδ or exported from the nucleus. Using FGF1 mutants with reduced binding to nucleolin and a FGF1-phosphomimetic mutant, we showed that the nucleolin-FGF1 interaction is critical for the intranuclear phosphorylation of FGF1 by PKCδ and thereby the regulation of nuclear export of FGF1.
Collapse
Affiliation(s)
- Torunn Sletten
- Department of Biochemistry, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Norway
| | - Michal Kostas
- Department of Protein Biotechnology, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Joanna Bober
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Vigdis Sorensen
- Department of Biochemistry, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Norway
| | - Mandana Yadollahi
- Department of Biochemistry, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Norway
| | - Sjur Olsnes
- Department of Biochemistry, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Norway
| | - Justyna Tomala
- Department of Protein Biotechnology, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Jacek Otlewski
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Malgorzata Zakrzewska
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Antoni Wiedlocha
- Department of Biochemistry, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Norway
| |
Collapse
|
30
|
The effects of p38 gene silencing on breast cancer cells. Mol Biol Rep 2014; 41:2923-7. [PMID: 24464183 DOI: 10.1007/s11033-014-3148-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 01/13/2014] [Indexed: 12/13/2022]
Abstract
p38 mitogen-activated protein kinases (MAPK) are members of the MAPK family that are activated by inflammatory cytokines and a variety of environmental stresses. It mediates various biological processes. p38 MAPK activity play important roles in tumour progression. Excessive p38 expression is observed in invasive breast cancers. The aim of the present study was to investigate whether the p38 siRNA transfection of breast cancer cells is a putative preventive treatment for human breast cancer. p38 siRNA was used at a concentration of 15, 30, and 100 nM in human breast cancer cell lines (MCF-7) and normal fibroblast cell lines (NIH 3T3). After 48 and 72 h of transfection, the reduction in p38 expression was measured using quantitative real-time PCR. The activation of p38 signalling was measured by ELISA. XTT cell proliferation assay was performed to determine the effect of p38 silencing on MCF-7 and NIH 3T3 cell lines. The results demonstrated that approximately 96% gene silencing occurred by the selected siRNA targeting p38 mRNA. The most effective silencing was observed at 72 h post-transfection using 30 nM p38 siRNA. The results of ELISA showed that the expression of p38 protein was inhibited by p38 siRNA at 30 nM siRNA and 100 nM at 72 h post transfection. XTT results showed that cells stimulated by 30 nM siRNA at 72 h post transfection were the lowest in proliferation. p38 siRNA can interfere with the expression of p38 at protein level in MCF-7 cells, result in inhibition of cell proliferation. p38 siRNA may be a critical regulator to promote the proliferation and protein expression in MCF-7 cells. In this study, we demonstrate that p38 silencing is a preventive maintenance for treating breast cancer.
Collapse
|
31
|
RSK2 regulates endocytosis of FGF receptor 1 by phosphorylation on serine 789. Oncogene 2013; 33:4823-36. [DOI: 10.1038/onc.2013.425] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 08/29/2013] [Accepted: 09/09/2013] [Indexed: 12/13/2022]
|
32
|
Fahrer J, Rausch J, Barth H. A cell-permeable fusion protein based on Clostridium botulinum C2 toxin for delivery of p53 tumorsuppressor into cancer cells. PLoS One 2013; 8:e72455. [PMID: 24039769 PMCID: PMC3764140 DOI: 10.1371/journal.pone.0072455] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2013] [Accepted: 07/18/2013] [Indexed: 01/09/2023] Open
Abstract
Genetically engineered bacterial protein toxins are attractive systems for delivery of exogenous proteins into the cytosol of mammalian cells. The binary C2 toxin from C. botulinum has emerged as powerful delivery vehicle, which rests on its binding/translocation component C2IIa and the genetically modified adaptor domain C2IN that act in concert to trigger cell uptake. The p53 tumor suppressor protein has a crucial function in suppressing carcinogenesis and is frequently inactivated by diverse mechanisms in human tumor cells. Therefore, we constructed a C2IN-p53 fusion protein, which is internalized into cancer cells by C2IIa. To this end, the C2IN-p53 fusion construct was overexpressed in E. coli with good solubility, purified by heparin affinity chromatography and protein identity was confirmed by immunoblotting. We demonstrated that the fusion protein is capable of binding to the p53 consensus-DNA with high affinity in a p53-specific manner in vitro. Next, the internalization of C2IN-p53 was monitored in HeLa cells by cell fractionation and immunoblot analysis, which revealed a C2IIa-mediated translocation of the fusion protein into the cytosol. The uptake was also shown in A549 and Saos-2 cells with similar efficiency. These findings were further corroborated by confocal immunofluorescence analyses of C2IN-p53/C2IIa-treated HeLa and A549 cells, displaying predominantly cytoplasmic localization of the fusion construct.
Collapse
Affiliation(s)
- Jörg Fahrer
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Ulm, Germany ; Institute of Toxicology, University Medical Center Mainz, Mainz, Germany
| | | | | |
Collapse
|
33
|
Son TW, Yun SP, Yong MS, Seo BN, Ryu JM, Youn HY, Oh YM, Han HJ. Netrin-1 protects hypoxia-induced mitochondrial apoptosis through HSP27 expression via DCC- and integrin α6β4-dependent Akt, GSK-3β, and HSF-1 in mesenchymal stem cells. Cell Death Dis 2013; 4:e563. [PMID: 23538444 PMCID: PMC3615739 DOI: 10.1038/cddis.2013.94] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Netrin (Ntn) has the potential to be successfully applied as an anti-apoptotic agent with a high affinity for tissue, for therapeutic strategies of umbilical cord blood-derived mesenchymal stem cells (UCB-MSC), although the mechanism by which Ntn-1 protects hypoxic injury has yet to be identified. Therefore, the present study examined the effect of Ntn-1 on hypoxia-induced UCB-MSC apoptosis, as well as the potential underlying mechanisms of its protective effect. Hypoxia (72 h) reduced cell viability (MTT reduction, and [3H]-thymidine incorporation) and cell number, and induced apoptosis (annexin and/or PI positive), which were reversed by Ntn-1 (10 ng/ml). Moreover, Ntn-1 decreased the increase of hypoxia-induced Bax, cleaved caspase-9, and -3, but blocked the decrease of hypoxia-reduced Bcl-2. Next, in order to examine the Ntn-1-related signaling cascade in the protection of hypoxic injury, we analyzed six Ntn receptors in UCB-MSC. We identified deleted in colorectal cancer (DCC) and integrin (IN) α6β4, except uncoordinated family member (UNC) 5A–C, and neogenin. Among them, IN α6β4 only was detected in lipid raft fractions. In addition, Ntn-1 induced the dissociation of DCC and APPL-1 complex, thereby stimulating the formation of APPL-1 and Akt2 complex. Ntn-1 also reversed the hypoxia-induced decrease of Akt and glycogen synthase kinase 3β (GSK-3β) phosphorylation, which is involved in heat shock factor-1 (HSF-1) expression. Ntn-1-induced phospho-Akt and -GSK-3β were inhibited by DCC function-blocking antibody, IN a6b4 function-blocking antibody, and the Akt inhibitor. Hypoxia and/or Ntn-1 stimulated heat shock protein (HSP)27 expression, which was blocked by HSF-1-specific small interfering RNA (siRNA). Furthermore, HSP27-specific siRNA reversed the Ntn-1-induced increase of phospho-Akt. Additionally, HSP27-specific siRNA attenuated the Ntn-1-reduced loss of mitochondrial membrane injury via the inhibition of cytochrome c (cyt c) release and formation of cyt c and HSP27 complex. Moreover, the inhibition of each signaling protein attenuated Ntn-1-induced blockage of apoptosis. In conclusion, Ntn-1-induced HSP27 protected hypoxic injury-related UCB-MSC apoptosis through DCC- and IN α6β4-dependent Akt, GSK-3β, and HSF-1 signaling pathways.
Collapse
Affiliation(s)
- T W Son
- Department of Veterinary Internal Medicine, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Fahrer J, Schweitzer B, Fiedler K, Langer T, Gierschik P, Barth H. C2-streptavidin mediates the delivery of biotin-conjugated tumor suppressor protein p53 into tumor cells. Bioconjug Chem 2013; 24:595-603. [PMID: 23506195 DOI: 10.1021/bc300563c] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We have previously generated a recombinant C2-streptavidin fusion protein for the delivery of biotin-labeled molecules of low molecular weight into the cytosol of mammalian cells. A nontoxic moiety of Clostridium botulinum C2 toxin mediates the cellular uptake, whereas the streptavidin unit serves as a binding platform for biotin-labeled cargo molecules. In the present study, we used the C2-streptavidin transporter to introduce biotin-conjugated p53 protein into various mammalian cell lines. The p53 tumor suppressor protein is inactivated in many human cancers by multiple mechanisms and therefore the restoration of its activity in tumor cells is of great therapeutic interest. Recombinant p53 was expressed in insect cells and biotin-labeled. Biotin-p53 retained its specific high-affinity DNA-binding as revealed by gel-shift analysis. Successful conjugation of biotin-p53 to the C2-streptavidin transporter was monitored by an overlay blot technique and confirmed by real-time surface plasmon resonance, providing a KD-value in the low nM range. C2-streptavidin significantly enhanced the uptake of biotin-p53 into African Green Monkey (Vero) epithelial cells as shown by flow cytometry. Using cell fractionation, the cytosolic translocation of biotin-p53 was detected in Vero cells as well as in HeLa cervix carcinoma cells. In line with this finding, confocal microscopy displayed cytoplasmic staining of biotin-p53 in HeLa and HL60 leukemia cells. Internalized biotin-p53 partially colocalized with early endosomes, as confirmed by confocal microscopy. In conclusion, our results demonstrate the successful conjugation of biotin-p53 to C2-streptavidin and its subsequent receptor-mediated endocytosis into different human tumor cell lines.
Collapse
Affiliation(s)
- Jörg Fahrer
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Albert-Einstein-Allee 11, D-89081 Ulm, Germany
| | | | | | | | | | | |
Collapse
|
35
|
Zakrzewska M, Haugsten EM, Nadratowska-Wesolowska B, Oppelt A, Hausott B, Jin Y, Otlewski J, Wesche J, Wiedlocha A. ERK-Mediated Phosphorylation of Fibroblast Growth Factor Receptor 1 on Ser777 Inhibits Signaling. Sci Signal 2013; 6:ra11. [DOI: 10.1126/scisignal.2003087] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
36
|
Yun SP, Ryu JM, Kim MO, Park JH, Han HJ. Rapid actions of plasma membrane estrogen receptors regulate motility of mouse embryonic stem cells through a profilin-1/cofilin-1-directed kinase signaling pathway. Mol Endocrinol 2012; 26:1291-303. [PMID: 22734041 DOI: 10.1210/me.2012-1002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Long-term estrogen actions are vital for driving cell growth, but more recent evidence suggests that estrogen mediates more rapid cellular effects. However, the function of estradiol-17β (E(2))-BSA in mouse embryonic stem cells has not been reported. Therefore, we examined the role of E(2)-BSA in mouse embryonic stem cell motility and its related signal pathways. E(2)-BSA (10(-8) m) significantly increased motility after 24 h incubation and increased filamentous (F)-actin expression; these effects were inhibited by the estrogen receptor antagonist ICI 182,780, indicating that E(2)-BSA bound membrane estrogen receptors and initiated a signal. E(2)-BSA increased c-Src and focal adhesion kinase (FAK) phosphorylation, which was attenuated by ICI 182,780. The E(2)-BSA-induced increase in epidermal growth factor receptor (EGFR) phosphorylation was inhibited by Src inhibitor PP2. As a downstream signal molecule, E(2)-BSA activated cdc42 and increased formation of a complex with the neural Wiskott-Aldrich syndrome protein (N-WASP)/cdc42/transducer of cdc42-dependent actin assembly-1 (TOCA-1), which was inhibited by FAK small interfering RNA (siRNA) and EGFR inhibitor AG 1478. In addition, E(2)-BSA increased profilin-1 expression and cofilin-1 phosphorylation, which was blocked by cdc42 siRNA. Subsequently, E(2)-BSA induced an increase in F-actin expression, and cell motility was inhibited by each signal pathway-related siRNA molecule or inhibitors but not by cofilin-1 siRNA. A combined treatment of cofilin-1 siRNA and E(2)-BSA increased F-actin expression and cell motility more than that of E(2)-BSA alone. These data demonstrate that E(2)-BSA stimulated motility by interacting with profilin-1/cofilin-1 and F-actin through FAK- and c-Src/EGFR transactivation-dependent N-WASP/cdc42/TOCA-1 complex.
Collapse
Affiliation(s)
- Seung Pil Yun
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju 500-757, Korea
| | | | | | | | | |
Collapse
|
37
|
Lillich M, Chen X, Weil T, Barth H, Fahrer J. Streptavidin-conjugated C3 protein mediates the delivery of mono-biotinylated RNAse A into macrophages. Bioconjug Chem 2012; 23:1426-36. [PMID: 22681511 DOI: 10.1021/bc300041z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The C3 toxin produced by Clostridium botulinum (C3bot) catalyzes the mono-ADP-ribosylation of the small GTPases Rho A, B and C, resulting in their inactivation. Recently, a specific endocytotic uptake mechanism of C3bot was identified in macrophages and myeloid leukemia cells. Here, we present a novel delivery system based upon a mutant C3bot devoid of ADP-ribosylation activity (C3Mut) and wild-type streptavidin (Stv). The C3Mut moiety mediates endocytosis into macrophages, whereas Stv functions as an adaptor protein for attaching biotinylated molecules to facilitate their subsequent internalization. First, a bioconjugate consisting of recombinant C3Mut and Stv was generated via a thioether linkage that tightly interacted with biotinylated bovine serum albumin as demonstrated by dot blot analysis. We then showed the internalization of C3Mut-Stv into J774A.1 macrophages by confocal microscopy and observed translocation into the cytosol using cell fractionation. The C3Mut-Stv bioconjugate did not affect cell viability. Next, we prepared mono-biotinylated RNase A, which was attached to the C3Mut-Stv transporter, and demonstrated its C3Mut-Stv-mediated delivery into the cytosol of J774A.1 cells. Finally, C3Mut-Stv also promoted the efficient uptake of mono-biotinylated lysozyme into J774A.1 cells, highlighting its versatility. This study intriguingly demonstrates the use of the novel C3Mut-Stv delivery system for protein transduction and may provide a basis for future applications, in particular, of cytotoxic RNase A mutants.
Collapse
Affiliation(s)
- Maren Lillich
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Albert-Einstein-Allee 11, D-89081 Ulm, Germany
| | | | | | | | | |
Collapse
|
38
|
Zhen Y, Sørensen V, Skjerpen CS, Haugsten EM, Jin Y, Wälchli S, Olsnes S, Wiedlocha A. Nuclear Import of Exogenous FGF1 Requires the ER-Protein LRRC59 and the Importins Kpnα1 and Kpnβ1. Traffic 2012; 13:650-64. [DOI: 10.1111/j.1600-0854.2012.01341.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Revised: 02/07/2012] [Accepted: 02/09/2012] [Indexed: 01/19/2023]
Affiliation(s)
| | | | | | | | | | - Sebastien Wälchli
- Department of Immunology; Institute for Cancer Research; The Norwegian Radium Hospital; Montebello; Oslo; 0310; Norway
| | | | | |
Collapse
|
39
|
Azad N, Iyer AKV, Wang L, Liu Y, Lu Y, Rojanasakul Y. Reactive oxygen species-mediated p38 MAPK regulates carbon nanotube-induced fibrogenic and angiogenic responses. Nanotoxicology 2012; 7:157-68. [PMID: 22263913 DOI: 10.3109/17435390.2011.647929] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Single-walled carbon nanotubes (SWCNTs) are fibrous nanoparticles that are being used widely for various applications including drug delivery. SWCNTs are currently under special attention for possible cytotoxicity. Recent reports suggest that exposure to nanoparticles leads to pulmonary fibrosis. We report that SWCNT-mediated interplay of fibrogenic and angiogenic regulators leads to increased angiogenesis, which is a novel finding that furthers the understanding of SWCNT-induced cytotoxicity. SWCNTs induce fibrogenesis through reactive oxygen species-regulated phosphorylation of p38 mitogen-activated protein kinase (MAPK). Activation of p38 MAPK by SWCNTs led to the induction of transforming growth factor (TGF)-β1 as well as vascular endothelial growth factor (VEGF). Both TGF-β1 and VEGF contributed significantly to the fibroproliferative and collagen-inducing effects of SWCNTs. Interestingly, a positive feedback loop was observed between TGF-β1 and VEGF. This interplay of fibrogenic and angiogenic mediators led to increased angiogenesis in response to SWCNTs. Overall this study reveals key signalling molecules involved in SWCNT-induced fibrogenesis and angiogenesis.
Collapse
Affiliation(s)
- Neelam Azad
- Department of Pharmaceutical Sciences, Hampton University, Hampton, VA 23668, USA
| | | | | | | | | | | |
Collapse
|
40
|
He L, Hristova K. Physical-chemical principles underlying RTK activation, and their implications for human disease. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2011; 1818:995-1005. [PMID: 21840295 DOI: 10.1016/j.bbamem.2011.07.044] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Revised: 07/27/2011] [Accepted: 07/29/2011] [Indexed: 12/15/2022]
Abstract
RTKs, the second largest family of membrane receptors, exert control over cell proliferation, differentiation and migration. In recent years, our understanding of RTK structure and activation in health and disease has skyrocketed. Here we describe experimental approaches used to interrogate RTKs, and we review the quantitative biophysical frameworks and structural considerations that shape our understanding of RTK function. We discuss current knowledge about RTK interactions, focusing on the role of different domains in RTK homodimerization, and on the importance and challenges in RTK heterodimerization studies. We also review our understanding of pathogenic RTK mutations, and the underlying physical-chemical causes for the pathologies. This article is part of a Special Issue entitled: Protein Folding in Membranes.
Collapse
Affiliation(s)
- Lijuan He
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | | |
Collapse
|
41
|
Zakrzewska M, Sørensen V, Jin Y, Wiedlocha A, Olsnes S. Translocation of exogenous FGF1 into cytosol and nucleus is a periodic event independent of receptor kinase activity. Exp Cell Res 2011; 317:1005-15. [DOI: 10.1016/j.yexcr.2011.01.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Revised: 12/10/2010] [Accepted: 01/03/2011] [Indexed: 10/18/2022]
|
42
|
Fahrer J, Funk J, Lillich M, Barth H. Internalization of biotinylated compounds into cancer cells is promoted by a molecular Trojan horse based upon core streptavidin and clostridial C2 toxin. Naunyn Schmiedebergs Arch Pharmacol 2010; 383:263-73. [DOI: 10.1007/s00210-010-0585-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2010] [Accepted: 11/24/2010] [Indexed: 12/16/2022]
|
43
|
Fahrer J, Rieger J, van Zandbergen G, Barth H. The C2-streptavidin delivery system promotes the uptake of biotinylated molecules in macrophages and T-leukemia cells. Biol Chem 2010; 391:1315-25. [DOI: 10.1515/bc.2010.132] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
AbstractMacrophages are tightly associated with inflammatory diseases as well as carcinogenesis, and therefore represent promising targets for drug delivery and gene transfer. We have recently established a novel protein delivery system based on the binary C2 toxin ofClostridium botulinumand streptavidin, allowing the uptake of exogenous biotinylated molecules into mammalian cells. Here, we applied this C2-streptavidin delivery system to macrophages and other leukocytes. First, the effect of wild-type C2 toxin on different leukocyte cell lines was tested, indicating no differences in sensitivity. Next, the uptake and stability of the engineered C2-streptavidin was analyzed in macrophages and Jurkat T-cells, showing internalization into the cytosol of both cell types with similar kinetics. The transporter did not exhibit any cytotoxic effect and did not interfere with phagocytosis in primary human macrophages. The C2-streptavidin system promoted specific uptake of biotinylated fluorophores into the cytosol of macrophages as revealed by confocal microscopy. In addition, flow cytometry analysis showed a significantly enhanced uptake of biotinylated fluorescent tracers in Jurkat leukemia cells mediated by the C2-streptavidin transporter. Our results demonstrate that C2-streptavidin is a functional delivery system for transport of biotinylated molecules into macrophages and other leukocytes without compromising cell viability and intrinsic functions such as phagocytosis.
Collapse
|
44
|
Abstract
The p38 MAPK (mitogen-activated protein kinase) signalling pathway allows cells to interpret a wide range of external signals and respond appropriately by generating a plethora of different biological effects. The diversity and specificity in cellular outcomes is achieved with an apparently simple linear architecture of the pathway, consisting of a core of three protein kinases acting sequentially. In the present review, we dissect the molecular mechanisms underlying p38 MAPK functions, with special emphasis on the activation and regulation of the core kinases, the interplay with other signalling pathways and the nature of p38 MAPK substrates as a source of functional diversity. Finally, we discuss how genetic mouse models are facilitating the identification of physiological functions for p38 MAPKs, which may impinge on their eventual use as therapeutic targets.
Collapse
|
45
|
Fahrer JÃ, Kuban J, Heine K, Rupps G, Kaiser E, Felder E, Benz R, Barth H. Selective and specific internalization of clostridial C3 ADP-ribosyltransferases into macrophages and monocytes. Cell Microbiol 2010; 12:233-47. [DOI: 10.1111/j.1462-5822.2009.01393.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
46
|
Fahrer J, Plunien R, Binder U, Langer T, Seliger H, Barth H. Genetically Engineered Clostridial C2 Toxin as a Novel Delivery System for Living Mammalian Cells. Bioconjug Chem 2009; 21:130-9. [DOI: 10.1021/bc900365b] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Jörg Fahrer
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Albert-Einstein-Allee 11, and Research Group on Chemical Functions in Biosystems, University of Ulm, Albert-Einstein-Allee 47, D-89081 Ulm, Germany
| | - Rainer Plunien
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Albert-Einstein-Allee 11, and Research Group on Chemical Functions in Biosystems, University of Ulm, Albert-Einstein-Allee 47, D-89081 Ulm, Germany
| | - Ulrike Binder
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Albert-Einstein-Allee 11, and Research Group on Chemical Functions in Biosystems, University of Ulm, Albert-Einstein-Allee 47, D-89081 Ulm, Germany
| | - Torben Langer
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Albert-Einstein-Allee 11, and Research Group on Chemical Functions in Biosystems, University of Ulm, Albert-Einstein-Allee 47, D-89081 Ulm, Germany
| | - Hartmut Seliger
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Albert-Einstein-Allee 11, and Research Group on Chemical Functions in Biosystems, University of Ulm, Albert-Einstein-Allee 47, D-89081 Ulm, Germany
| | - Holger Barth
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Albert-Einstein-Allee 11, and Research Group on Chemical Functions in Biosystems, University of Ulm, Albert-Einstein-Allee 47, D-89081 Ulm, Germany
| |
Collapse
|
47
|
Zakrzewska M, Wiedlocha A, Szlachcic A, Krowarsch D, Otlewski J, Olsnes S. Increased protein stability of FGF1 can compensate for its reduced affinity for heparin. J Biol Chem 2009; 284:25388-403. [PMID: 19574212 PMCID: PMC2757240 DOI: 10.1074/jbc.m109.001289] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Human FGF1 (fibroblast growth factor 1) is a powerful signaling molecule with a short half-life in vivo and a denaturation temperature close to physiological. Binding to heparin increases the stability of FGF1 and is believed to be important in the formation of FGF1.fibroblast growth factor receptor (FGFR) active complex. In order to reveal the function of heparin in FGF1.FGFR complex formation and signaling, we constructed several FGF1 variants with reduced affinity for heparin and with diverse stability. We determined their biophysical properties and biological activities as well as their ability to translocate across cellular membranes. Our study showed that increased thermodynamic stability of FGF1 nicely compensates for decreased binding of heparin in FGFR activation, induction of DNA synthesis, and cell proliferation. By stepwise introduction of stabilizing mutations into the K118E (K132E) FGF1 variant that shows reduced affinity for heparin and is inactive in stimulation of DNA synthesis, we were able to restore the full mitogenic activity of this mutant. Our results indicate that the main role of heparin in FGF-induced signaling is to protect this naturally unstable protein against heat and/or proteolytic degradation and that heparin is not essential for a direct FGF1-FGFR interaction and receptor activation.
Collapse
Affiliation(s)
- Malgorzata Zakrzewska
- Centre for Cancer Biomedicine, University of Oslo, and Department of Biochemistry, Institute for Cancer Research, Norwegian Radium Hospital, Montebello, 0310 Oslo, Norway
| | | | | | | | | | | |
Collapse
|
48
|
Naidu S, Vijayan V, Santoso S, Kietzmann T, Immenschuh S. Inhibition and genetic deficiency of p38 MAPK up-regulates heme oxygenase-1 gene expression via Nrf2. THE JOURNAL OF IMMUNOLOGY 2009; 182:7048-57. [PMID: 19454702 DOI: 10.4049/jimmunol.0900006] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Heme oxygenase (HO)-1 is the inducible isoform of the first and rate-limiting enzyme of heme degradation. The HO products carbon monoxide and bilirubin not only provide antioxidant cytoprotection, but also have potent anti-inflammatory and immunomodulatory functions. Although HO-1 has previously been shown to be induced by various stimuli via activation of the p38 MAPK signaling pathway, the role of this protein kinase for HO-1 gene regulation is largely unknown. In the present study, it is demonstrated that pharmacological inhibitors of p38 induced HO-1 expression in monocytic cells. Moreover, basal HO-1 gene expression levels were markedly higher in untreated murine embryonic fibroblasts (MEF) from p38alpha(-/-) mice compared with those from wild-type mice. Transfection studies with luciferase reporter gene constructs indicate that increased HO-1 gene expression via inhibition of p38 was mediated by the transcription factor Nrf2, which is a central regulator of the cellular oxidative stress response. Accordingly, inhibitors of p38 induced binding of nuclear proteins to a Nrf2 target sequence of the HO-1 promoter, but did not affect HO-1 protein expression and promoter activity in Nrf2(-/-) MEF. Genetic deficiency of p38 led to enhanced phosphorylation of ERK and increased cellular accumulation of reactive oxygen species. In addition, pharmacological blockage of ERK and scavenging of reactive oxygen species with N-acetylcysteine reduced HO-1 gene expression in p38(-/-) MEF, respectively. Taken together, it is demonstrated that pharmacological inhibition and genetic deficiency of p38 induce HO-1 gene expression via a Nrf2-dependent mechanism in monocytic cells and MEF.
Collapse
Affiliation(s)
- Srivatsava Naidu
- Institute for Clinical Immunology and Transfusion Medicine, Justus-Liebig-University Giessen, Giessen, Germany
| | | | | | | | | |
Collapse
|
49
|
The protein phosphatase 2A regulatory subunits B'beta and B'delta mediate sustained TrkA neurotrophin receptor autophosphorylation and neuronal differentiation. Mol Cell Biol 2008; 29:662-74. [PMID: 19029245 DOI: 10.1128/mcb.01242-08] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Nerve growth factor (NGF) is critical for the differentiation and maintenance of neurons in the peripheral and central nervous system. Sustained autophosphorylation of the TrkA receptor tyrosine kinase and long-lasting activation of downstream kinase cascades are hallmarks of NGF signaling, yet our knowledge of the molecular mechanisms underlying prolonged TrkA activity is incomplete. Protein phosphatase 2A (PP2A) is a heterotrimeric Ser/Thr phosphatase composed of a scaffolding, catalytic, and regulatory subunit (B, B', and B" gene families). Here, we employ a combination of pharmacological inhibitors, regulatory subunit overexpression, PP2A scaffold subunit exchange, and RNA interference to show that PP2A containing B' family regulatory subunits participates in sustained NGF signaling in PC12 cells. Specifically, two neuron-enriched regulatory subunits, B'beta and B'delta, recruit PP2A into a complex with TrkA to dephosphorylate the NGF receptor on Ser/Thr residues and to potentiate its intrinsic Tyr kinase activity. Acting at the receptor level, PP2A/ B'beta and B'delta enhance NGF (but not epidermal growth factor or fibroblast growth factor) signaling through the Akt and Ras-mitogen-activated protein kinase cascades and promote neuritogenesis and differentiation of PC12 cells. Thus, select PP2A heterotrimers oppose desensitization of the TrkA receptor tyrosine kinase, perhaps through dephosphorylation of inhibitory Ser/Thr phosphorylation sites on the receptor itself, to maintain neurotrophin-mediated developmental and survival signaling.
Collapse
|