1
|
Shimada N, Matsuda J, Asano-Matsuda K, Tokuchi M, Aoudjit L, Masztalerz A, Lemay S, Takano T, Isaka Y. Rac1 Suppression by the Focal Adhesion Protein GIT ArfGAP2 and Podocyte Protection. J Am Soc Nephrol 2025:00001751-990000000-00532. [PMID: 40019803 DOI: 10.1681/asn.0000000614] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 01/15/2025] [Indexed: 04/30/2025] Open
Abstract
Key Points
Focal adhesion protein GIT2 protected podocytes from injury in rodent proteinuric disease models.GIT2 facilitated translocation of tyrosine phosphatase PTP1B to focal adhesions where it dephosphorylates p130Cas, thereby suppressing Rac1 activity.Stabilizing GIT2 or facilitating GIT2 localization to focal adhesions in podocytes could be a therapeutic strategy in proteinuric kidney diseases.
Background
Podocytes have an intricate structure featured by numerous actin-based projections called foot processes. Rho family of small GTPases, including Ras-related C3 botulinum toxin substrate 1 (Rac1), play important roles in actin cytoskeletal remodeling required for cell morphology and adhesion. We previously showed that Rac1 activation in podocytes causes foot process effacement and proteinuria, but the upstream and spatiotemporal regulatory mechanism directing Rac1 is largely unknown. Recently, we identified the focal adhesion protein GIT ArfGAP2 (GIT2) as one of the Rac1 interactors in human podocytes by proximity-dependent biotin identification and proteomics.
Methods
Systemic and podocyte-specific GIT2 knockout mice were generated and assessed for kidney phenotypes. Human podocytes with GIT2 knockdown (KD) and overexpression were established using lentiviral transduction and characterized.
Results
GIT2 was enriched in glomeruli, including podocytes, in the mouse kidney. Gene deletion of Git2 in podocytes caused exacerbated proteinuria and foot process effacement when subjected to the minimal change disease model and salt-sensitive hypertension model, which were improved by pharmacological inhibition of Rac1. In cultured podocytes, GIT2 KD resulted in Rac1-dependent cell spreading with marked lamellipodial protrusions, accelerated focal adhesion disassembly, and shorter focal adhesion lifetime. In GIT2 KD podocytes, tyrosine phosphorylation of the focal adhesion protein p130 Crk-associated substrate (Cas) was significantly increased, accompanied by impaired localization of the tyrosine phosphatase PTP1B to focal adhesions. These phenotypes observed in GIT2 KD podocytes were reversed by GIT2 overexpression.
Conclusions
The results indicate that GIT2 facilitates translocation of PTP1B to focal adhesions where it dephosphorylates p130Cas, thereby suppressing local Rac1 activity and protecting against podocyte injury and proteinuria.
Collapse
Affiliation(s)
- Naoyuki Shimada
- Department of Nephrology, Graduate School of Medicine, The University of Osaka, Suita, Osaka, Japan
| | - Jun Matsuda
- Department of Nephrology, Graduate School of Medicine, The University of Osaka, Suita, Osaka, Japan
| | - Kana Asano-Matsuda
- Department of Nephrology, Graduate School of Medicine, The University of Osaka, Suita, Osaka, Japan
| | - Maho Tokuchi
- Department of Nephrology, Graduate School of Medicine, The University of Osaka, Suita, Osaka, Japan
| | - Lamine Aoudjit
- Division of Nephrology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Agnieszka Masztalerz
- Division of Nephrology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Serge Lemay
- Division of Nephrology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Tomoko Takano
- Division of Nephrology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Yoshitaka Isaka
- Department of Nephrology, Graduate School of Medicine, The University of Osaka, Suita, Osaka, Japan
| |
Collapse
|
2
|
Giese MA, Bennin DA, Schoen TJ, Peterson AN, Schrope JH, Brand J, Jung HS, Keller NP, Beebe DJ, Dinh HQ, Slukvin II, Huttenlocher A. PTP1B phosphatase dampens iPSC-derived neutrophil motility and antimicrobial function. J Leukoc Biol 2024; 116:118-131. [PMID: 38417030 PMCID: PMC11212797 DOI: 10.1093/jleuko/qiae039] [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: 08/25/2023] [Revised: 02/09/2024] [Accepted: 02/12/2024] [Indexed: 03/01/2024] Open
Abstract
Neutrophils are rapidly recruited to sites of infection and are critical for pathogen clearance. Therapeutic use of primary neutrophils has been limited, as they have a short lifespan and are not amenable to genetic manipulation. Human induced pluripotent stem cells (iPSCs) can provide a robust source of neutrophils for infusion and are genetically tractable. However, current work has indicated that dampened intracellular signaling limits iPSC-derived neutrophil (iNeutrophil) cellular activation and antimicrobial response. Here, we show that protein tyrosine phosphatase 1B (PTP1B) inhibits intracellular signaling and dampens iNeutrophil effector function. Deletion of the PTP1B phosphatase increased PI3K and ERK signaling and was associated with increased F-actin polymerization, cell migration, and phagocytosis. In contrast, other effector functions like NETosis and reactive oxygen species production were reduced. PTP1B-deficient neutrophils were more responsive to Aspergillus fumigatus and displayed rapid recruitment and control of hyphal growth. Accordingly, depletion of PTP1B increased production of inflammatory factors including the neutrophil chemokine interleukin-8. Taken together, these findings suggest that PTP1B limits iNeutrophil motility and antimicrobial function.
Collapse
Affiliation(s)
- Morgan A Giese
- Department of Medical Microbiology and Immunology, University of Wisconsin–Madison, 1550 Linden Dr. Madison 53706, WI, United States
- Cellular and Molecular Biology Graduate Program, University of Wisconsin–Madison, 1525 Linden Dr. Madison 53706, WI, United States
| | - David A Bennin
- Department of Medical Microbiology and Immunology, University of Wisconsin–Madison, 1550 Linden Dr. Madison 53706, WI, United States
| | - Taylor J Schoen
- Department of Medical Microbiology and Immunology, University of Wisconsin–Madison, 1550 Linden Dr. Madison 53706, WI, United States
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin–Madison, 2015 Linden Dr. Madison 53706, WI, United States
| | - Ashley N Peterson
- Department of Medical Microbiology and Immunology, University of Wisconsin–Madison, 1550 Linden Dr. Madison 53706, WI, United States
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin–Madison, 2015 Linden Dr. Madison 53706, WI, United States
| | - Jonathan H Schrope
- Department of Biomedical Engineering, University of Wisconsin–Madison, 1550 Engineering Dr. Madison 53706, WI, United States
| | - Josh Brand
- Cell and Molecular Pathology Graduate Program, University of Wisconsin–Madison, 1685 Highland Ave. Madison 53705, WI, United States
- Department of Oncology, McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin–Madison, 1111 Highland Ave. Madison 53705, WI, United States
| | - Ho Sun Jung
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Ct. Madison 53715, WI, United States
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, 1111 Highland Ave. Madison 53705, WI, United States
| | - Nancy P Keller
- Department of Medical Microbiology and Immunology, University of Wisconsin–Madison, 1550 Linden Dr. Madison 53706, WI, United States
| | - David J Beebe
- Carbone Cancer Center, University of Wisconsin–Madison, 1111 Highland Ave. Madison 53705, WI, United States
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, 1685 Highland Ave. Madison 53705, WI, United States
| | - Huy Q Dinh
- Department of Oncology, McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin–Madison, 1111 Highland Ave. Madison 53705, WI, United States
| | - Igor I Slukvin
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Ct. Madison 53715, WI, United States
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, 1111 Highland Ave. Madison 53705, WI, United States
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, 1685 Highland Ave. Madison 53705, WI, United States
| | - Anna Huttenlocher
- Department of Medical Microbiology and Immunology, University of Wisconsin–Madison, 1550 Linden Dr. Madison 53706, WI, United States
- Department of Pediatrics, University of Wisconsin–Madison, 600 Highland Ave. Madison 53705, WI, United States
| |
Collapse
|
3
|
Samaržija I, Konjevoda P. Extracellular Matrix- and Integrin Adhesion Complexes-Related Genes in the Prognosis of Prostate Cancer Patients' Progression-Free Survival. Biomedicines 2023; 11:2006. [PMID: 37509645 PMCID: PMC10377098 DOI: 10.3390/biomedicines11072006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/12/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Prostate cancer is a heterogeneous disease, and one of the main obstacles in its management is the inability to foresee its course. Therefore, novel biomarkers are needed that will guide the treatment options. The extracellular matrix (ECM) is an important part of the tumor microenvironment that largely influences cell behavior. ECM components are ligands for integrin receptors which are involved in every step of tumor progression. An underlying characteristic of integrin activation and ligation is the formation of integrin adhesion complexes (IACs), intracellular structures that carry information conveyed by integrins. By using The Cancer Genome Atlas data, we show that the expression of ECM- and IACs-related genes is changed in prostate cancer. Moreover, machine learning methods revealed that they are a source of biomarkers for progression-free survival of patients that are stratified according to the Gleason score. Namely, low expression of FMOD and high expression of PTPN2 genes are associated with worse survival of patients with a Gleason score lower than 9. The FMOD gene encodes protein that may play a role in the assembly of the ECM and the PTPN2 gene product is a protein tyrosine phosphatase activated by integrins. Our results suggest potential biomarkers of prostate cancer progression.
Collapse
Affiliation(s)
- Ivana Samaržija
- Laboratory for Epigenomics, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Paško Konjevoda
- Laboratory for Epigenomics, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| |
Collapse
|
4
|
Saadeldin IM, Tanga BM, Bang S, Seo C, Koo O, Yun SH, Kim SI, Lee S, Cho J. ROCK Inhibitor (Y-27632) Abolishes the Negative Impacts of miR-155 in the Endometrium-Derived Extracellular Vesicles and Supports Embryo Attachment. Cells 2022; 11:cells11193178. [PMID: 36231141 PMCID: PMC9564368 DOI: 10.3390/cells11193178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/07/2022] [Accepted: 10/07/2022] [Indexed: 11/16/2022] Open
Abstract
Extracellular vesicles (EVs) are nanosized vesicles that act as snapshots of cellular components and mediate cellular communications, but they may contain cargo contents with undesired effects. We developed a model to improve the effects of endometrium-derived EVs (Endo-EVs) on the porcine embryo attachment in feeder-free culture conditions. Endo-EVs cargo contents were analyzed using conventional and real-time PCR for micro-RNAs, messenger RNAs, and proteomics. Porcine embryos were generated by parthenogenetic electric activation in feeder-free culture conditions supplemented with or without Endo-EVs. The cellular uptake of Endo-EVs was confirmed using the lipophilic dye PKH26. Endo-EVs cargo contained miR-100, miR-132, and miR-155, together with the mRNAs of porcine endogenous retrovirus (PERV) and β-catenin. Targeting PERV with CRISPR/Cas9 resulted in reduced expression of PERV mRNA transcripts and increased miR-155 in the Endo-EVs, and supplementing these in embryos reduced embryo attachment. Supplementing the medium containing Endo-EVs with miR-155 inhibitor significantly improved the embryo attachment with a few outgrowths, while supplementing with Rho-kinase inhibitor (RI, Y-27632) dramatically improved both embryo attachment and outgrowths. Moreover, the expression of miR-100, miR-132, and the mRNA transcripts of BCL2, zinc finger E-box-binding homeobox 1, β-catenin, interferon-γ, protein tyrosine phosphatase non-receptor type 1, PERV, and cyclin-dependent kinase 2 were all increased in embryos supplemented with Endo-EVs + RI compared to those in the control group. Endo-EVs + RI reduced apoptosis and increased the expression of OCT4 and CDX2 and the cell number of embryonic outgrowths. We examined the individual and combined effects of RI compared to those of the miR-155 mimic and found that RI can alleviate the negative effects of the miR-155 mimic on embryo attachment and outgrowths. EVs can improve embryo attachment and the unwanted effects of the de trop cargo contents (miR-155) can be alleviated through anti-apoptotic molecules such as the ROCK inhibitor.
Collapse
Affiliation(s)
- Islam M. Saadeldin
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
- Research Institute of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
| | - Bereket Molla Tanga
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
| | - Seonggyu Bang
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
| | - Chaerim Seo
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
| | | | - Sung Ho Yun
- Korea Basic Science Institute (KBSI), Ochang 28119, Korea
| | - Seung Il Kim
- Korea Basic Science Institute (KBSI), Ochang 28119, Korea
| | - Sanghoon Lee
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
| | - Jongki Cho
- Laboratory of Theriogenology, College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
- Correspondence: ; Tel.: +82-42-821-6788
| |
Collapse
|
5
|
Uprety B, Abrahamse H. Targeting Breast Cancer and Their Stem Cell Population through AMPK Activation: Novel Insights. Cells 2022; 11:576. [PMID: 35159385 PMCID: PMC8834477 DOI: 10.3390/cells11030576] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 02/06/2023] Open
Abstract
Despite some significant advancements, breast cancer has become the most prevalent cancer in the world. One of the main reasons for failure in treatment and metastasis has been attributed to the presence of cancer initiating cells-cancer stem cells. Consequently, research is now being focussed on targeting cancer cells along with their stem cell population. Non-oncology drugs are gaining increasing attention for their potent anticancer activities. Metformin, a drug commonly used to treat type 2 diabetes, is the best example in this regard. It exerts its therapeutic action by activating 5' adenosine monophosphate-activated protein kinase (AMPK). Activated AMPK subsequently phosphorylates and targets several cellular pathways involved in cell growth and proliferation and the maintenance of stem-like properties of cancer stem cells. Therefore, AMPK is emerging as a target of choice for developing effective anticancer drugs. Vanadium compounds are well-known PTP inhibitors and AMPK activators. They find extensive applications in treatment of diabetes and obesity via PTP1B inhibition and AMPK-mediated inhibition of adipogenesis. However, their role in targeting cancer stem cells has not been explored yet. This review is an attempt to establish the applications of insulin mimetic vanadium compounds for the treatment of breast cancer by AMPK activation and PTP1B inhibition pathways.
Collapse
Affiliation(s)
- Bhawna Uprety
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa;
| | | |
Collapse
|
6
|
González Wusener AE, González Á, Perez Collado ME, Maza MR, General IJ, Arregui CO. Protein tyrosine phosphatase 1B targets focal adhesion kinase and paxillin in cell-matrix adhesions. J Cell Sci 2021; 134:272564. [PMID: 34553765 DOI: 10.1242/jcs.258769] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 09/14/2021] [Indexed: 11/20/2022] Open
Abstract
Protein tyrosine phosphatase 1B (PTP1B, also known as PTPN1) is an established regulator of cell-matrix adhesion and motility. However, the nature of substrate targets at adhesion sites remains to be validated. Here, we used bimolecular fluorescence complementation assays, in combination with a substrate trapping mutant of PTP1B, to directly examine whether relevant phosphotyrosines on paxillin and focal adhesion kinase (FAK, also known as PTK2) are substrates of the phosphatase in the context of cell-matrix adhesion sites. We found that the formation of catalytic complexes at cell-matrix adhesions requires intact tyrosine residues Y31 and Y118 on paxillin, and the localization of FAK at adhesion sites. Additionally, we found that PTP1B specifically targets Y925 on the focal adhesion targeting (FAT) domain of FAK at adhesion sites. Electrostatic analysis indicated that dephosphorylation of this residue promotes the closed conformation of the FAT 4-helix bundle and its interaction with paxillin at adhesion sites.
Collapse
Affiliation(s)
- Ana E González Wusener
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires 1650, Argentina
| | - Ángela González
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires 1650, Argentina
| | - María E Perez Collado
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires 1650, Argentina
| | - Melina R Maza
- Escuela de Ciencia y Tecnología, Universidad Nacional de San Martin, Instituto de Ciencias Físicas and CONICET, San Martin, Buenos Aires 1650, Argentina
| | - Ignacio J General
- Escuela de Ciencia y Tecnología, Universidad Nacional de San Martin, Instituto de Ciencias Físicas and CONICET, San Martin, Buenos Aires 1650, Argentina
| | - Carlos O Arregui
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires 1650, Argentina
| |
Collapse
|
7
|
Rashmi, More SK, Wang Q, Vomhof‐DeKrey EE, Porter JE, Basson MD. ZINC40099027 activates human focal adhesion kinase by accelerating the enzymatic activity of the FAK kinase domain. Pharmacol Res Perspect 2021; 9:e00737. [PMID: 33715263 PMCID: PMC7955952 DOI: 10.1002/prp2.737] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 01/28/2021] [Accepted: 01/29/2021] [Indexed: 12/16/2022] Open
Abstract
Focal adhesion kinase (FAK) regulates gastrointestinal epithelial restitution and healing. ZINC40099027 (Zn27) activates cellular FAK and promotes intestinal epithelial wound closure in vitro and in mice. However, whether Zn27 activates FAK directly or indirectly remains unknown. We evaluated Zn27 potential modulation of the key phosphatases, PTP-PEST, PTP1B, and SHP2, that inactivate FAK, and performed in vitro kinase assays with purified FAK to assess direct Zn27-FAK interaction. In human Caco-2 cells, Zn27-stimulated FAK-Tyr-397 phosphorylation despite PTP-PEST inhibition and did not affect PTP1B-FAK interaction or SHP2 activity. Conversely, in vitro kinase assays demonstrated that Zn27 directly activates both full-length 125 kDa FAK and its 35 kDa kinase domain. The ATP-competitive FAK inhibitor PF573228 reduced basal and ZN27-stimulated FAK phosphorylation in Caco-2 cells, but Zn27 increased FAK phosphorylation even in cells treated with PF573228. Increasing PF573228 concentrations completely prevented activation of 35 kDa FAK in vitro by a normally effective Zn27 concentration. Conversely, increasing Zn27 concentrations dose-dependently activated kinase activity and overcame PF573228 inhibition of FAK, suggesting the direct interactions of Zn27 with FAK may be competitive. Zn27 increased the maximal activity (Vmax ) of FAK. The apparent Km of the substrate also increased under laboratory conditions less relevant to intracellular ATP concentrations. These results suggest that Zn27 is highly potent and enhances FAK activity via allosteric interaction with the FAK kinase domain to increase the Vmax of FAK for ATP. Understanding Zn27 enhancement of FAK activity will be important to redesign and develop a clinical drug that can promote mucosal wound healing.
Collapse
Affiliation(s)
- Rashmi
- Department of SurgeryUniversity of North Dakota School of Medicine & Health SciencesGrand ForksNDUSA
| | - Shyam K. More
- Department of SurgeryUniversity of North Dakota School of Medicine & Health SciencesGrand ForksNDUSA
| | - Qinggang Wang
- Department of SurgeryUniversity of North Dakota School of Medicine & Health SciencesGrand ForksNDUSA
| | - Emilie E. Vomhof‐DeKrey
- Department of SurgeryUniversity of North Dakota School of Medicine & Health SciencesGrand ForksNDUSA
- Department of Biomedical SciencesUniversity of North Dakota School of Medicine & Health SciencesGrand ForksNDUSA
| | - James E. Porter
- Department of Biomedical SciencesUniversity of North Dakota School of Medicine & Health SciencesGrand ForksNDUSA
| | - Marc D. Basson
- Department of SurgeryUniversity of North Dakota School of Medicine & Health SciencesGrand ForksNDUSA
- Department of Biomedical SciencesUniversity of North Dakota School of Medicine & Health SciencesGrand ForksNDUSA
- Department of PathologyUniversity of North Dakota School of Medicine & Health SciencesGrand ForksNDUSA
| |
Collapse
|
8
|
Tang Y, Luo K, Chen Y, Chen Y, Zhou R, Chen C, Tan J, Deng M, Dai Q, Yu X, Liu J, Zhang C, Wu W, Xu J, Dong S, Luo F. Phosphorylation inhibition of protein-tyrosine phosphatase 1B tyrosine-152 induces bone regeneration coupled with angiogenesis for bone tissue engineering. Bioact Mater 2021; 6:2039-2057. [PMID: 33511306 PMCID: PMC7809253 DOI: 10.1016/j.bioactmat.2020.12.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 12/06/2020] [Accepted: 12/25/2020] [Indexed: 12/26/2022] Open
Abstract
A close relationship has been reported to exist between cadherin-mediated cell–cell adhesion and integrin-mediated cell mobility, and protein tyrosine phosphatase 1B (PTP1B) may be involved in maintaining this homeostasis. The stable residence of mesenchymal stem cells (MSCs) and endothelial cells (ECs) in their niches is closely related to the regulation of PTP1B. However, the exact role of the departure of MSCs and ECs from their niches during bone regeneration is largely unknown. Here, we show that the phosphorylation state of PTP1B tyrosine-152 (Y152) plays a central role in initiating the departure of these cells from their niches and their subsequent recruitment to bone defects. Based on our previous design of a PTP1B Y152 region-mimicking peptide (152RM) that significantly inhibits the phosphorylation of PTP1B Y152, further investigations revealed that 152RM enhanced cell migration partly via integrin αvβ3 and promoted MSCs osteogenic differentiation partly by inhibiting ATF3. Moreover, 152RM induced type H vessels formation by activating Notch signaling. Demineralized bone matrix (DBM) scaffolds were fabricated with mesoporous silica nanoparticles (MSNs), and 152RM was then loaded onto them by electrostatic adsorption. The DBM-MSN/152RM scaffolds were demonstrated to induce bone formation and type H vessels expansion in vivo. In conclusion, our data reveal that 152RM contributes to bone formation by coupling osteogenesis with angiogenesis, which may offer a potential therapeutic strategy for bone defects. PTP1B plays a dual regulatory role in cadherin- and integrin-related pathways. Inhibition of PTP1B Y152 phosphorylation enhances the departure of MSCs from the stem cell niche. DBM-MSN/152RM scaffolds coordinate the recruitment of MSCs and ECs. DBM-MSN/152RM scaffolds promote bone regeneration and angiogenesis in bone defects.
Collapse
Affiliation(s)
- Yong Tang
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China.,Department of Orthopaedics, 72nd Group Army Hospital, Huzhou University, Huzhou, Zhejiang, China
| | - Keyu Luo
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China.,Department of Spine Surgery, Center for Orthopedics, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yin Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Yueqi Chen
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China.,Department of Biomedical Materials Science, Third Military Medical University, Chongqing, China
| | - Rui Zhou
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Can Chen
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jiulin Tan
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Moyuan Deng
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Qijie Dai
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xueke Yu
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jian Liu
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Chengmin Zhang
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Wenjie Wu
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jianzhong Xu
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Shiwu Dong
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China.,Department of Biomedical Materials Science, Third Military Medical University, Chongqing, China
| | - Fei Luo
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
9
|
McDermott MI, Wang Y, Wakelam MJO, Bankaitis VA. Mammalian phospholipase D: Function, and therapeutics. Prog Lipid Res 2019; 78:101018. [PMID: 31830503 DOI: 10.1016/j.plipres.2019.101018] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 10/08/2019] [Accepted: 10/14/2019] [Indexed: 01/23/2023]
Abstract
Despite being discovered over 60 years ago, the precise role of phospholipase D (PLD) is still being elucidated. PLD enzymes catalyze the hydrolysis of the phosphodiester bond of glycerophospholipids producing phosphatidic acid and the free headgroup. PLD family members are found in organisms ranging from viruses, and bacteria to plants, and mammals. They display a range of substrate specificities, are regulated by a diverse range of molecules, and have been implicated in a broad range of cellular processes including receptor signaling, cytoskeletal regulation and membrane trafficking. Recent technological advances including: the development of PLD knockout mice, isoform-specific antibodies, and specific inhibitors are finally permitting a thorough analysis of the in vivo role of mammalian PLDs. These studies are facilitating increased recognition of PLD's role in disease states including cancers and Alzheimer's disease, offering potential as a target for therapeutic intervention.
Collapse
Affiliation(s)
- M I McDermott
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843-1114, United States of America.
| | - Y Wang
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843-1114, United States of America; Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843-2128, United States of America
| | - M J O Wakelam
- Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, United Kingdom
| | - V A Bankaitis
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, TX 77843-1114, United States of America; Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843-2128, United States of America; Department of Chemistry, Texas A&M University, College Station, Texas 77840, United States of America
| |
Collapse
|
10
|
Ramos AR, Elong Edimo W, Erneux C. Phosphoinositide 5-phosphatase activities control cell motility in glioblastoma: Two phosphoinositides PI(4,5)P2 and PI(3,4)P2 are involved. Adv Biol Regul 2018; 67:40-48. [PMID: 28916189 DOI: 10.1016/j.jbior.2017.09.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 09/01/2017] [Accepted: 09/01/2017] [Indexed: 05/15/2023]
Abstract
Inositol polyphosphate 5-phosphatases or phosphoinositide 5-phosphatases (PI 5-phosphatases) are enzymes that can act on soluble inositol phosphates and/or phosphoinositides (PIs). Several PI 5-phosphatases have been linked to human genetic diseases, in particular the Lowe protein or OCRL which is mutated in the Lowe syndrome. There are 10 different members of this family and 9 of them can use PIs as substrate. One of these substrates, PI(3,4,5)P3 binds to specific PH domains and recruits as effectors specific proteins to signaling complexes. Protein kinase B is one target protein and activation of the kinase will have a major impact on cell proliferation, survival and cell metabolism. Two other PIs, PI(4,5)P2 and PI(3,4)P2, are produced or used as substrates of PI 5-phosphatases (OCRL, INPP5B, SHIP1/2, SYNJ1/2, INPP5K, INPP5J, INPP5E). The inositol lipids may influence many aspects of cytoskeletal organization, lamellipodia formation and F-actin polymerization. PI 5-phosphatases have been reported to control cell migration, adhesion, polarity and cell invasion particularly in cancer cells. In glioblastoma, reducing SHIP2 expression can positively or negatively affect the speed of cell migration depending on the glioblastoma cell type. The two PI 5-phosphatases SHIP2 or SKIP could be localized at the plasma membrane and can reduce either PI(3,4,5)P3 or PI(4,5)P2 abundance. In the glioblastoma 1321 N1 cells, SHIP2 controls plasma membrane PI(4,5)P2 thereby participating in the control of cell migration.
Collapse
Affiliation(s)
- Ana Raquel Ramos
- Interdisciplinary Research Institute (IRIBHM), Université Libre de Bruxelles, Campus Erasme, Bldg C, 808 Route de Lennik, 1070 Brussels, Belgium
| | - William's Elong Edimo
- Interdisciplinary Research Institute (IRIBHM), Université Libre de Bruxelles, Campus Erasme, Bldg C, 808 Route de Lennik, 1070 Brussels, Belgium
| | - Christophe Erneux
- Interdisciplinary Research Institute (IRIBHM), Université Libre de Bruxelles, Campus Erasme, Bldg C, 808 Route de Lennik, 1070 Brussels, Belgium.
| |
Collapse
|
11
|
Rossi MC, Bezerra FJB, Silva RA, Crulhas BP, Fernandes CJC, Nascimento AS, Pedrosa VA, Padilha P, Zambuzzi WF. Titanium-released from dental implant enhances pre-osteoblast adhesion by ROS modulating crucial intracellular pathways. J Biomed Mater Res A 2017. [PMID: 28639351 DOI: 10.1002/jbm.a.36150] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
It is important to understand the cellular and molecular events that occur at the cell-material interface of implants used for bone repair. The mechanisms involved in the initial stages of osteoblast interactions with the surface of the implant material must be decisive for cell fating surrounding them. In order to address this issue, we decided to investigate if conditioned medium for dental implants was able to modulate murine pre-osteoblast metabolism. First, we determined the concentration of titanium (Ti)-containing conditioned medium and found that it was 2-fold increased (p < 0.0001). We have reported that this conditioned medium significantly up-modulated pre-osteoblast adhesion up to 24 h (p < 0.0001). In parallel, our results showed that both phosphorylations of FAK (focal adhesion kinase) at Y397 (p < 0.0011) and Cofilin at Ser03 (p < 0.0053) were also up-modulated, as well as for Rac1 expression (p < 0.0175); both of them are involved with cell adaptation by rearranging cytoskeleton actin filaments. Thereafter, Ti-containing medium stimulated ROS (reactive oxygen species) production by pre-osteoblast cells, and it is very possible that ROS compromised PTP-1B (protein tyrosine phosphatase 1B) activation since PTP1B was down-phosphorylated (p < 0.0148). The low PTP activity guarantees the phosphorylation of FAK at Y-residue, causing better pre-osteoblast adhesion in response to Ti-containing medium. Altogether, these data indicate that ROS indirectly modulate FAK phosphorylation in response to Ti-released from dental implants. Taken the results in account, these data showed for the first time that the implanted dental device is able to dynamically affect surrounding tissues, mainly by promoting a better performance of the pre-osteoblast cells. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 2968-2976, 2017.
Collapse
Affiliation(s)
- M C Rossi
- Department of Chemistry and Biochemistry, Bioscience Institute, State University of São Paulo - UNESP, campus Botucatu, PO Box 510, Rubião Jr, Botucatu, São Paulo, 18618-970, Brazil
| | - F J B Bezerra
- Department of Chemistry and Biochemistry, Bioscience Institute, State University of São Paulo - UNESP, campus Botucatu, PO Box 510, Rubião Jr, Botucatu, São Paulo, 18618-970, Brazil
| | | | - B P Crulhas
- Department of Chemistry and Biochemistry, Bioscience Institute, State University of São Paulo - UNESP, campus Botucatu, PO Box 510, Rubião Jr, Botucatu, São Paulo, 18618-970, Brazil
| | - C J C Fernandes
- Department of Chemistry and Biochemistry, Bioscience Institute, State University of São Paulo - UNESP, campus Botucatu, PO Box 510, Rubião Jr, Botucatu, São Paulo, 18618-970, Brazil
| | - A S Nascimento
- Department of Chemistry and Biochemistry, Bioscience Institute, State University of São Paulo - UNESP, campus Botucatu, PO Box 510, Rubião Jr, Botucatu, São Paulo, 18618-970, Brazil
| | - V A Pedrosa
- Department of Chemistry and Biochemistry, Bioscience Institute, State University of São Paulo - UNESP, campus Botucatu, PO Box 510, Rubião Jr, Botucatu, São Paulo, 18618-970, Brazil
| | - P Padilha
- Department of Chemistry and Biochemistry, Bioscience Institute, State University of São Paulo - UNESP, campus Botucatu, PO Box 510, Rubião Jr, Botucatu, São Paulo, 18618-970, Brazil
| | - W F Zambuzzi
- Department of Chemistry and Biochemistry, Bioscience Institute, State University of São Paulo - UNESP, campus Botucatu, PO Box 510, Rubião Jr, Botucatu, São Paulo, 18618-970, Brazil
| |
Collapse
|
12
|
Wang Y, Yan F, Ye Q, Wu X, Jiang F. PTP1B inhibitor promotes endothelial cell motility by activating the DOCK180/Rac1 pathway. Sci Rep 2016; 6:24111. [PMID: 27052191 PMCID: PMC4823726 DOI: 10.1038/srep24111] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 03/21/2016] [Indexed: 12/14/2022] Open
Abstract
Promoting endothelial cell (EC) migration is important not only for therapeutic angiogenesis, but also for accelerating re-endothelialization after vessel injury. Several recent studies have shown that inhibition of protein tyrosine phosphatase 1B (PTP1B) may promote EC migration and angiogenesis by enhancing the vascular endothelial growth factor receptor-2 (VEGFR2) signalling. In the present study, we demonstrated that PTP1B inhibitor could promote EC adhesion, spreading and migration, which were abolished by the inhibitor of Rac1 but not RhoA GTPase. PTP1B inhibitor significantly increased phosphorylation of p130Cas, and the interactions among p130Cas, Crk and DOCK180; whereas the phosphorylation levels of focal adhesion kinase, Src, paxillin, or Vav2 were unchanged. Gene silencing of DOCK180, but not Vav2, abrogated the effects of PTP1B inhibitor on EC motility. The effects of PTP1B inhibitor on EC motility and p130Cas/DOCK180 activation persisted in the presence of the VEGFR2 antagonist. In conclusion, we suggest that stimulation of the DOCK180 pathway represents an alternative mechanism of PTP1B inhibitor-stimulated EC motility, which does not require concomitant VEGFR2 activation as a prerequisite. Therefore, PTP1B inhibitor may be a useful therapeutic strategy for promoting EC migration in cardiovascular patients in which the VEGF/VEGFR functions are compromised.
Collapse
Affiliation(s)
- Yuan Wang
- Key Laboratory of Cardiovascular Remodelling and Function Research (Chinese Ministry of Education and Chinese Ministry of Health) and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Feng Yan
- Key Laboratory of Cardiovascular Remodelling and Function Research (Chinese Ministry of Education and Chinese Ministry of Health) and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Qing Ye
- Key Laboratory of Cardiovascular Remodelling and Function Research (Chinese Ministry of Education and Chinese Ministry of Health) and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Xiao Wu
- Key Laboratory of Cardiovascular Remodelling and Function Research (Chinese Ministry of Education and Chinese Ministry of Health) and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Fan Jiang
- Key Laboratory of Cardiovascular Remodelling and Function Research (Chinese Ministry of Education and Chinese Ministry of Health) and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
13
|
González Wusener AE, González Á, Nakamura F, Arregui CO. PTP1B triggers integrin-mediated repression of myosin activity and modulates cell contractility. Biol Open 2015; 5:32-44. [PMID: 26700725 PMCID: PMC4728310 DOI: 10.1242/bio.015883] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cell contractility and migration by integrins depends on precise regulation of protein tyrosine kinase and Rho-family GTPase activities in specific spatiotemporal patterns. Here we show that protein tyrosine phosphatase PTP1B cooperates with β3 integrin to activate the Src/FAK signalling pathway which represses RhoA-myosin-dependent contractility. Using PTP1B null (KO) cells and PTP1B reconstituted (WT) cells, we determined that some early steps following cell adhesion to fibronectin and vitronectin occurred robustly in WT cells, including aggregation of β3 integrins and adaptor proteins, and activation of Src/FAK-dependent signalling at small puncta in a lamellipodium. However, these events were significantly impaired in KO cells. We established that cytoskeletal strain and cell contractility was highly enhanced at the periphery of KO cells compared to WT cells. Inhibition of the Src/FAK signalling pathway or expression of constitutive active RhoA in WT cells induced a KO cell phenotype. Conversely, expression of constitutive active Src or myosin inhibition in KO cells restored the WT phenotype. We propose that this novel function of PTP1B stimulates permissive conditions for adhesion and lamellipodium assembly at the protruding edge during cell spreading and migration. Summary: Here we show that protein tyrosine phosphatase PTP1B cooperates with β3 integrin to transiently repress RhoA-myosin-dependent contractility, stimulating adhesion and lamellipodium assembly during cell spreading and migration.
Collapse
Affiliation(s)
- Ana E González Wusener
- IIB-INTECH, Universidad Nacional de San Martín, 1650 San Martín, Buenos Aires, Argentina
| | - Ángela González
- IIB-INTECH, Universidad Nacional de San Martín, 1650 San Martín, Buenos Aires, Argentina
| | - Fumihiko Nakamura
- Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02445, USA
| | - Carlos O Arregui
- IIB-INTECH, Universidad Nacional de San Martín, 1650 San Martín, Buenos Aires, Argentina
| |
Collapse
|
14
|
Martin-Granados C, Prescott AR, Le Sommer S, Klaska IP, Yu T, Muckersie E, Giuraniuc CV, Grant L, Delibegovic M, Forrester JV. A key role for PTP1B in dendritic cell maturation, migration, and T cell activation. J Mol Cell Biol 2015; 7:517-28. [PMID: 26063615 DOI: 10.1093/jmcb/mjv032] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 03/08/2015] [Indexed: 12/15/2022] Open
Abstract
Dendritic cells (DC) are the major antigen-presenting cells bridging innate and adaptive immunity, a function they perform by converting quiescent DC to active, mature DC with the capacity to activate naïve T cells. They do this by migrating from the tissues to the T cell area of the secondary lymphoid tissues. Here, we demonstrate that myeloid cell-specific genetic deletion of PTP1B (LysM PTP1B) leads to defects in lipopolysaccharide-driven bone marrow-derived DC (BMDC) activation associated with increased levels of phosphorylated Stat3. We show that myeloid cell-specific PTP1B deletion also causes decreased migratory capacity of epidermal DC, as well as reduced CCR7 expression and chemotaxis to CCL19 by BMDC. PTP1B deficiency in BMDC also impairs their migration in vivo. Further, immature LysM PTP1B BMDC display fewer podosomes, increased levels of phosphorylated Src at tyrosine 527, and loss of Src localization to podosome puncta. In co-culture with T cells, LysM PTP1B BMDC establish fewer and shorter contacts than control BMDC. Finally, LysM PTP1B BMDC fail to present antigen to T cells as efficiently as control BMDC. These data provide first evidence for a key regulatory role for PTP1B in mediating a central DC function of initiating adaptive immune responses in response to innate immune cell activation.
Collapse
Affiliation(s)
- Cristina Martin-Granados
- Institute of Medical Sciences, University of Aberdeen, College of Life Sciences and Medicine, Aberdeen AB25 2ZD, UK Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, UK
| | - Alan R Prescott
- Division of Cell Biology and Immunology, College of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Samantha Le Sommer
- Institute of Medical Sciences, University of Aberdeen, College of Life Sciences and Medicine, Aberdeen AB25 2ZD, UK
| | - Izabela P Klaska
- Institute of Medical Sciences, University of Aberdeen, College of Life Sciences and Medicine, Aberdeen AB25 2ZD, UK
| | - Tian Yu
- Institute of Medical Sciences, University of Aberdeen, College of Life Sciences and Medicine, Aberdeen AB25 2ZD, UK
| | - Elizabeth Muckersie
- Institute of Medical Sciences, University of Aberdeen, College of Life Sciences and Medicine, Aberdeen AB25 2ZD, UK
| | - Claudiu V Giuraniuc
- Institute of Medical Sciences, University of Aberdeen, College of Life Sciences and Medicine, Aberdeen AB25 2ZD, UK
| | - Louise Grant
- Institute of Medical Sciences, University of Aberdeen, College of Life Sciences and Medicine, Aberdeen AB25 2ZD, UK
| | - Mirela Delibegovic
- Institute of Medical Sciences, University of Aberdeen, College of Life Sciences and Medicine, Aberdeen AB25 2ZD, UK
| | - John V Forrester
- Institute of Medical Sciences, University of Aberdeen, College of Life Sciences and Medicine, Aberdeen AB25 2ZD, UK Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, Australia Centre for Experimental Immunology, Lions Eye Institute, Nedlands, WA 6009, Australia
| |
Collapse
|
15
|
Mußbach F, Henklein P, Westermann M, Settmacher U, Böhmer FD, Kaufmann R. Proteinase-activated receptor 1- and 4-promoted migration of Hep3B hepatocellular carcinoma cells depends on ROS formation and RTK transactivation. J Cancer Res Clin Oncol 2015; 141:813-25. [PMID: 25373316 DOI: 10.1007/s00432-014-1863-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 10/22/2014] [Indexed: 02/08/2023]
Abstract
PURPOSE There is growing evidence for a role of proteinase-activated receptors (PARs), a subfamily of G protein-coupled receptors, in cancer. We have previously shown that PAR1 and PAR4 are able to promote the migration of hepatocellular carcinoma (HCC) cells suggesting a function in HCC progression. In this study, we assessed the underlying signalling mechanisms. METHODS Using Hep3B liver carcinoma cells, RTK activation was assessed by Western blot employing phospho-RTK specific antibodies, ROS level were estimated by H2DCF-DA using confocal laser scanning microscopy, and measurement of PTP activity was performed in cell lysates using 6,8-difluoro-4-methylumbelliferyl phosphate (DiFMUP) as a substrate. RESULTS Thrombin, the PAR1 selective agonist peptide TFLLRN-NH2 (PAR1-AP), and the PAR4 selective agonist peptide, AYPGKF-NH2 (PAR4-AP), induced a significant increase in Hep3B cell migration that could be blocked by inhibitors targeting formation of reactive oxygen species (ROS), or activation of hepatocyte-growth factor receptor (Met), or platelet-derived growth factor receptor (PDGFR), respectively. The involvement of these intracellular effectors in PAR1/4-initiated migratory signalling was further supported by the findings that individual stimulation of Hep3B cells with the PAR1-AP and the PAR4-AP induced an increase in ROS production and the transactivation of Met and PDGFR. In addition, PAR1- and PAR4-mediated inhibition of total PTP activity and specifically PTP1B. ROS inhibition by N-acetyl-L-cysteine prevented the inhibition of PTP1B phosphatase activity induced by PAR1-AP and the PAR4-AP, but had no effect on PAR1/4-mediated activation of Met and PDGFR in Hep3B cells. CONCLUSIONS Collectively, our data indicate that PAR1 and PAR4 activate common promigratory signalling pathways in Hep3B liver carcinoma cells including activation of the receptor tyrosine kinases Met and PDGFR, the formation of ROS and the inactivation of PTP1B. However, PAR1/4-triggered Met and PDGFR transactivation seem to be mediated independently from the ROS-PTP1B signalling module.
Collapse
Affiliation(s)
- Franziska Mußbach
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, Erlanger Allee 101, 07747, Jena, Germany
| | | | | | | | | | | |
Collapse
|
16
|
Sjoelund V, Smelkinson M, Nita-Lazar A. Phosphoproteome profiling of the macrophage response to different toll-like receptor ligands identifies differences in global phosphorylation dynamics. J Proteome Res 2014; 13:5185-97. [PMID: 24941444 PMCID: PMC4227906 DOI: 10.1021/pr5002466] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
![]()
Toll-like
receptors (TLRs) are among the first sensors that detect
infection and drive immune response. Macrophages encountering a pathogen
are usually stimulated not by one TLR, but by a combination of TLRs
engaged by distinct microbe ligands. To understand the integrated
signaling under complex conditions, we investigated the differences
in the phosphoprotein signaling cascades triggered by TLR2, TLR4,
and TLR7 ligands using a single responding cell population. We performed
a global, quantitative, early poststimulation kinetic analysis of
the mouse macrophage phosphoproteome using stable isotope labeling
with amino acids coupled to phosphopeptide enrichment and high-resolution
mass spectrometry. For each TLR ligand, we found marked elevation
of phosphorylation of cytoskeleton components, GTPases of the Rho
family, and phospholipase C signaling pathway proteins. Phosphorylation
of proteins involved in phagocytosis was only seen in response to
TLR2 and TLR4 but not to TLR7 activation. Changes in the phosphorylation
of proteins involved in endocytosis were delayed in response to TLR2
as compared to TLR4 ligands. These findings reveal that the phosphoproteomic
response to stimulation of distinct TLRs varies both in the major
modification targets and the phosphorylation dynamics. These results
advance the understanding of how macrophages sense and respond to
a diverse set of TLR stimuli.
Collapse
Affiliation(s)
- Virginie Sjoelund
- Laboratory of Systems Biology, and §Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, Maryland 20892, United States
| | | | | |
Collapse
|