1
|
He P, Wang Z, Yang J, Pan P, Shi T, Xu S, Lan J, Hao Z, Yang A, Chen L, Xi Y, Wang J. Mechanism of Ligusticum wallichii-Borneol in the Treatment of Cerebral Ischemic Stroke in Rats Based On Network Pharmacology, Molecular Docking, and Experimental Verification. Chem Biodivers 2025; 22:e202401893. [PMID: 39714965 DOI: 10.1002/cbdv.202401893] [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: 08/09/2024] [Revised: 12/15/2024] [Accepted: 12/18/2024] [Indexed: 12/25/2024]
Abstract
The pharmacodynamics, molecular biology, network pharmacology, and molecular docking mechanisms of the Ligusticum wallichii and borneol medication pair (CXBP) were investigated for ischemic stroke treatment. Effective chemical components and targets of CXBP were identified using TCMSP, ETCM, and SymMap databases, whereas ischemic stroke targets were sourced from OMIM, GeneCards, TTD, PubMed, Web of Science, CNKI, Wanfang Data, and VIP databases. Protein-protein interaction (PPI) networks were generated using the STRING database, and GO and KEGG enrichment analyses were conducted using Metascape. A "disease-pathway-target-component-drug" network was created in Cytoscape, and molecular docking was confirmed with PyMOL and AutoDock tools. Rat models of MCAO were established to evaluate neurological scores, triphenyltetrazolium chloride (TTC) staining, and Nissl staining. Key components were validated through enzyme-linked immunosorbent assay (ELISA), real-time polymerase chain reaction (PCR), and immunohistochemistry. Thirty-three active ingredients and 419 potential targets for CXBP, with key compounds including Z-6,8',7,3'-diligustilide, cedrene, (+)-alpha-funebrene, POL, dipterocarpol, oleanolic acid, 1-acetyl-beta-carboline, and erythrodiol. Critical targets included ESR1, PRKCA, and PTPN6. KEGG pathway analysis revealed 179 signaling pathways, primarily neuroactive ligand-receptor interactions, whereas GO enrichment analysis identified 2911 biological processes, 398 molecular activities, and 203 cellular components. The neurological function scores and TTC staining of the infarcted brain regions were significantly improved following CXBP intervention compared to the MCAO group. These findings were supported by Nissl staining, which demonstrated better preserved cellular morphology and a significantly higher number of Nissl vesicles in the CXBP group. ELISA analysis revealed substantial modulation in gene expression: levels of PRKCA, PTPN6, ESR1, and TNF-α changed significantly, whereas IL-1β, IL-6, and TNF-α were notably downregulated compared to the MCAO group. PCR results corroborated these findings, showing a significant decrease in PRKCA expression and marked downregulation of IL-1β, IL-6, and TNF-α, whereas ESR1 and PTPN6 levels increased significantly. Immunohistochemical analysis further confirmed these results, with the CXBP and nimodipine groups exhibiting higher ESR1 and PTPN6 expression and lower PRKCA expression compared to the MCAO group. To improve cerebral ischemia and reperfusion injury, CXBP improves ischemic stroke outcomes through key active ingredients (e.g., Z-6,8',7,3'-diligustilide, cedrene, and oleanolic acid) and targets ESR1, PRKCA, and PTPN6, modulating multiple signaling pathways to alleviate cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Pengfen He
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Zhifeng Wang
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Jiao Yang
- Second Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Pan Pan
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Ting Shi
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Shuangfeng Xu
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Junfeng Lan
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Zhihui Hao
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Aiming Yang
- Department of Neurology, Yunnan Provincial Hospital of Chinese Medicine, Kunming, Yunnan, China
| | - Liang Chen
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Yujiang Xi
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Jian Wang
- Department of Neurology, First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| |
Collapse
|
2
|
Yu L, Gu X, Chen P, Yang R, Xu Y, Yang X. Effects of PTPN6 Gene Knockdown in SKM-1 Cells on Apoptosis, Erythroid Differentiation and Inflammations. Curr Issues Mol Biol 2024; 46:12061-12074. [PMID: 39590309 PMCID: PMC11593023 DOI: 10.3390/cimb46110715] [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: 08/20/2024] [Revised: 10/06/2024] [Accepted: 10/25/2024] [Indexed: 11/28/2024] Open
Abstract
Objective: Protein tyrosine phosphatase non-receptor type 6 (PTPN6) is a cytoplasmic phosphatase that acts as a key regulatory protein in cell signaling to control inflammation and cell death. In order to investigate the role of PTPN6 in hematologic tumor myelodysplastic syndrome (MDS), this study infected SKM-1 cell line (MDS cell line) with packaged H_PTPN6-shRNA lentivirus to obtain H_PTPN6-shRNA SKM-1 stable strain. The effect of PTPN6 knockdown on apoptosis, erythroid differentiation, and inflammations in SKM-1 cell line was examined. Methods: The stable knockdown SKM-1 cell line was validated using qPCR and Western blot assays. The proliferation activity, apoptosi, erythroid differentiation, and inflammatory cytokines in SKM-1 cells were assessed before and after transfection. Results: qPCR confirmed that the expression level of H_PTPN6-shRNA in SKM-1 cells was significantly reduced, and Western blot showed that the protein expression level of H_PTPN6-shRNA in SKM-1 cells was also significantly reduced. The CCK-8 cell viability assay confirmed that stable gene knockdown did not affect cell viability. Flow cytometry revealed that the apoptosis rate of cells in the PTPN6 knockdown group was 0.8%, lower than the 2.7% observed in the empty plasmid group; the expression rate of the erythroid differentiation marker CD235a was 13.2%, lower than the 25.0% observed in the empty plasmid group. The expression levels of the proinflammatory factors IL-6 and IL-8 increased, and the expression levels of the inhibitor factor IL-4 decreased. Conclusions: The PTPN6 gene was successfully knocked down using lentivirus-mediated transduction, and the constructed cell line was validated using PCR and Western blot. The CCK-8 cell viability assay confirmed that stable gene knockdown did not affect cell proliferation viability. Flow cytometry analysis of apoptosis and erythroid differentiation indicated that PTPN6 knockdown inhibits apoptosis and erythroid differentiation in SKM-1 cells and also alters the level of inflammations in the bone marrow microenvironment. It suggests that the PTPN6 gene acts as a tumor suppressor in myelodysplastic syndrome cells, influencing hematopoietic cell apoptosis, erythroid differentiation, and inflammations. This provides a reliable experimental basis for further in-depth studies on the mechanism of PTPN6 in MDS and related pharmacological research.
Collapse
Affiliation(s)
| | | | | | | | - Yonggang Xu
- Department of Hematology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; (L.Y.); (X.G.); (P.C.); (R.Y.)
| | - Xiupeng Yang
- Department of Hematology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; (L.Y.); (X.G.); (P.C.); (R.Y.)
| |
Collapse
|
3
|
Bhasin S, Das A. Marine alkaloid rigidin analogues as potential selective inhibitors of SHP1, a new strategy for cancer immunotherapeutics. J Biomol Struct Dyn 2024; 42:5590-5606. [PMID: 37349914 DOI: 10.1080/07391102.2023.2227708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/14/2023] [Indexed: 06/24/2023]
Abstract
SHP1 is a protein tyrosine phosphatase playing a central role in immunity, cell growth, development, and survival. The inhibition of SHP1 can help in better prognosis in various disorders like breast and ovarian cancer, melanoma, atherosclerosis, hypoxia, hypoactive immune response, and familial dysautonomia. The currently available inhibitors of SHP1 have the side effect of inhibiting the activity of SHP2, which shares >60% sequence similarity with SHP1 but has distinct biological functions. Thus, there is a need to search for novel specific inhibitors of SHP1. The current study uses a combination of virtual screening and molecular dynamic simulations, followed by PCA and MM-GBSA analysis, to screen about 35000 compounds; to predict that two rigidin analogues can potentially selectively inhibit SHP1 but not SHP2. Our studies demonstrate that these rigidin analogues are more potent at inhibiting SHP1 than the commercially available inhibitor NSC-87877. Further, cross-binding studies with SHP2 exhibited poor binding efficiency and lower stability of the complex, thus indicating a specificity of the rigidin analogues for SHP1, which is crucial in preventing side effects due to the diverse physiological functions of SHP2 in cellular signaling, proliferation, and hematopoiesis. Additionally, SHP1 is essential in mediating the inhibitory signaling in antitumor immune cells like NK and T cells. Hence, the rigidin analogues that inhibit SHP1 will potentiate the anti-tumor immune response by the release of inhibitory function of NK cells, thus driving NK activating response, in addition to their intrinsic anti-tumor function. Thus, SHP1 inhibition is a novel double-blade approach towards anti-cancer immunotherapeutics.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sidharth Bhasin
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
- Department of Biotechnology, Delhi Technological University, Shahbad Daulatpur, Delhi, India
| | - Asmita Das
- Department of Biotechnology, Delhi Technological University, Shahbad Daulatpur, Delhi, India
| |
Collapse
|
4
|
Mey M, Bhatta S, Suresh S, Labrador LM, Piontkivska H, Casadesus G. Therapeutic benefits of central LH receptor agonism in the APP/PS1 AD model involve trophic and immune regulation and are reproductive status dependent. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167165. [PMID: 38653355 DOI: 10.1016/j.bbadis.2024.167165] [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: 02/06/2024] [Revised: 04/02/2024] [Accepted: 04/08/2024] [Indexed: 04/25/2024]
Abstract
The mechanisms that underly reproductive hormone effects on cognition, neuronal plasticity, and AD risk, particularly in relation to gonadotropin LH receptor (LHCGR) signaling, remain poorly understood. To address this gap in knowledge and clarify the impact of circulating steroid hormones on the therapeutic effects of CNS LHCGR activation, we delivered the LHCGR agonist human chorionic gonadotropin (hCG) intracerebroventricularly (ICV) and evaluated functional, structural, plasticity-related signaling cascades, Aβ pathology, and transcriptome differences in reproductively intact and ovariectomized (OVX) APP/PS1 AD female mice. Here we demonstrate that CNS hCG delivery restored function to wild-type levels only in OVX APP/PS1 mice. Spine density was increased in all hCG treated groups independently of reproductive status. Notably, increases in BDNF signaling and cognition, were selectively upregulated only in the OVX hCG-treated group. RNA sequencing analyses identified a significant increase in peripheral myeloid and pro-inflammatory genes within the hippocampi of the OVX group that were completely reversed by hCG treatment, identifying a potential mechanism underlying the selective therapeutic effect of LHCGR activation. Interestingly, in intact mice, hCG administration mimicked the effects of gonadectomy. Together, our findings indicate that CNS LHCGR agonism in the post-menopausal context is beneficial through trophic and immune mechanisms. Our findings also underscore the presence of a steroid-LHCGR mechanistic interaction that is unexplored yet potentially meaningful to fully understand "post-menopausal" brain function and CNS hormone treatment response.
Collapse
Affiliation(s)
- Megan Mey
- Kent State University, Kent, OH 44240, United States of America
| | - Sabina Bhatta
- Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Sneha Suresh
- University of Florida, Gainesville, FL 32606, United States of America
| | | | | | - Gemma Casadesus
- University of Florida, Gainesville, FL 32606, United States of America.
| |
Collapse
|
5
|
Mey M, Bhatta S, Suresh S, Montero Labrador L, Piontkivska H, Casadesus G. The LH receptor regulates hippocampal spatial memory and restores dendritic spine density in ovariectomized APP/PS1 AD mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.22.573087. [PMID: 38187770 PMCID: PMC10769359 DOI: 10.1101/2023.12.22.573087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Activation of the luteinizing hormone receptor (LHCGR) rescues spatial memory function and spine density losses associated with gonadectomy and high circulating gonadotropin levels in females. However, whether this extends to the AD brain or the mechanisms that underlie these benefits remain unknown. To address this question, we delivered the LHCGR agonist human chorionic gonadotropin (hCG) intracerebroventricularly (ICV), under reproductively intact and ovariectomized conditions to mimic the post-menopausal state in the APP/PS1mouse brain. Cognitive function was tested using the Morris water maze task, and hippocampal dendritic spine density, Aβ pathology, and signaling changes associated with these endpoints were determined to address mechanisms. Here we show that central LHCGR activation restored function in ovariectomized APP/PS1 female mice to wild-type levels without altering Aβ pathology. LHCGR activation increased hippocampal dendritic spine density regardless of reproductive status, and this was mediated by BDNF-dependent and independent signaling. We also show that ovariectomy in the APP/PS1 brain elicits an increase in peripherally derived pro-inflammatory genes which are inhibited by LHCGR activation. This may mediate reproductive status specific effects of LHCGR agonism on cognitive function and BDNF expression. Together, this work highlights the relevance of the LHCGR on cognition and its therapeutic potential in the "menopausal" AD brain.
Collapse
|
6
|
Yarnall MT, Kim SH, Korntner S, Bishop AC. Destabilization of the SHP2 and SHP1 protein tyrosine phosphatase domains by a non-conserved "backdoor" cysteine. Biochem Biophys Rep 2022; 32:101370. [PMID: 36275931 PMCID: PMC9578986 DOI: 10.1016/j.bbrep.2022.101370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 10/11/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022] Open
Abstract
Protein tyrosine phosphatases (PTPs) are critical regulators of cellular signal transduction that catalyze the hydrolytic dephosphorylation of phosphotyrosine in substrate proteins. Among several conserved features in classical PTP domains are an active-site cysteine residue that is necessary for catalysis and a "backdoor" cysteine residue that can serve to protect the active-site cysteine from irreversible oxidation. Curiously, two biologically important phosphatases, Src homology domain-containing PTPs 2 and 1 (SHP2 and SHP1), each contain an additional backdoor cysteine residue at a position of the PTP domain that is occupied by proline in almost all other classical PTPs (position 333 in human SHP2 numbering). Here we show that the presence of cysteine 333 significantly destabilizes the fold of the PTP domains in the SHPs. We find that replacement of cysteine 333 with proline confers increased thermal stability on the SHP2 and SHP1 PTP domains, as measured by temperature-dependent activity assays and differential scanning fluorimetry. Conversely, we show that substantial destabilization of the PTP-domain fold is conferred by introduction of a non-natural cysteine residue in a non-SHP PTP that contains proline at the 333 position. It has previously been suggested that the extra backdoor cysteine of the SHP PTPs may work in tandem with the conserved backdoor cysteine to provide protection from irreversible oxidative enzyme inactivation. If so, our current results suggest that, during the course of mammalian evolution, the SHP proteins have developed extra protection from oxidation at the cost of the thermal instability that is conferred by the presence of their PTP domains' second backdoor cysteine.
Collapse
Affiliation(s)
| | - Sean H. Kim
- Amherst College, Department of Chemistry, Amherst, MA, 01002, USA
| | - Samuel Korntner
- Amherst College, Department of Chemistry, Amherst, MA, 01002, USA
| | | |
Collapse
|
7
|
Kang Q, Li L, Pang Y, Zhu W, Meng L. An update on Ym1 and its immunoregulatory role in diseases. Front Immunol 2022; 13:891220. [PMID: 35967383 PMCID: PMC9366555 DOI: 10.3389/fimmu.2022.891220] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 07/06/2022] [Indexed: 11/23/2022] Open
Abstract
Ym1 is a rodent-specific chitinase-like protein (CLP) lacking catalytic activity, whose cellular origins are mainly macrophages, neutrophils and other cells. Although the detailed function of Ym1 remains poorly understood, Ym1 has been generally recognized as a fundamental feature of alternative activation of macrophages in mice and hence one of the prevalent detecting targets in macrophage phenotype distinguishment. Studies have pointed out that Ym1 may have regulatory effects, which are multifaceted and even contradictory, far more than just a mere marker. Allergic lung inflammation, parasite infection, autoimmune diseases, and central nervous system diseases have been found associations with Ym1 to varying degrees. Thus, insights into Ym1’s role in diseases would help us understand the pathogenesis of different diseases and clarify the genuine roles of CLPs in mammals. This review summarizes the information on Ym1 from the gene to its expression and regulation and focuses on the association between Ym1 and diseases.
Collapse
Affiliation(s)
- Qi Kang
- Institute of Molecular and Translational Medicine, and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi’an Jiaotong University, Ministry of Education, Xi’an, China
- Department of Clinical Medicine, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Luyao Li
- Institute of Molecular and Translational Medicine, and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi’an Jiaotong University, Ministry of Education, Xi’an, China
- Department of Clinical Medicine, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Yucheng Pang
- Institute of Molecular and Translational Medicine, and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi’an Jiaotong University, Ministry of Education, Xi’an, China
- Department of Clinical Medicine, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Wenhua Zhu
- Institute of Molecular and Translational Medicine, and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi’an Jiaotong University, Ministry of Education, Xi’an, China
- *Correspondence: Wenhua Zhu, ; Liesu Meng,
| | - Liesu Meng
- Institute of Molecular and Translational Medicine, and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi’an Jiaotong University, Ministry of Education, Xi’an, China
- National Joint Engineering Research Center of Biodiagnostics and Biotherapy, Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Wenhua Zhu, ; Liesu Meng,
| |
Collapse
|
8
|
Zhang Q, Kong W, Zhu T, Zhu G, Zhu J, Kong X, Du Y. Phase separation ability and phosphatase activity of the SHP1-R360E mutant. Biochem Biophys Res Commun 2022; 600:150-155. [DOI: 10.1016/j.bbrc.2022.02.070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 02/18/2022] [Indexed: 11/15/2022]
|
9
|
Boncompagni G, Varone A, Tatangelo V, Capitani N, Frezzato F, Visentin A, Trentin L, Corda D, Baldari CT, Patrussi L. Glycerophosphoinositol Promotes Apoptosis of Chronic Lymphocytic Leukemia Cells by Enhancing Bax Expression and Activation. Front Oncol 2022; 12:835290. [PMID: 35392232 PMCID: PMC8980805 DOI: 10.3389/fonc.2022.835290] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
An imbalance in the expression of pro- and anti-apoptotic members of the Bcl-2 family of apoptosis-regulating proteins is one of the main biological features of CLL, highlighting these proteins as therapeutic targets for treatment of this malignancy. Indeed, the Bcl-2 inhibitor Venetoclax is currently used for both first-line treatment and treatment of relapsed or refractory CLL. An alternative avenue is the transcriptional modulation of Bcl-2 family members to tilt their balance towards apoptosis. Glycerophosphoinositol (GroPIns) is a biomolecule generated from membrane phosphoinositides by the enzymes phospholipase A2 and lysolipase that pleiotropically affects key cellular functions. Mass-spectrometry analysis of GroPIns interactors recently highlighted the ability of GroPIns to bind to the non-receptor tyrosine phosphatase SHP-1, a known promoter of Bax expression, suggesting that GroPIns might correct the Bax expression defect in CLL cells, thereby promoting their apoptotic demise. To test this hypothesis, we cultured CLL cells in the presence of GroPIns, alone or in combination with drugs commonly used for treatment of CLL. We found that GroPIns alone increases Bax expression and apoptosis in CLL cells and enhances the pro-apoptotic activity of drugs used for CLL treatment in a SHP-1 dependent manner. Interestingly, among GroPIns interactors we found Bax itself. Short-term treatments of CLL cells with GroPIns induce Bax activation and translocation to the mitochondria. Moreover, GroPIns enhances the pro-apoptotic activity of Venetoclax and Fludarabine in CLL cells. These data provide evidence that GroPIns exploits two different pathways converging on Bax to promote apoptosis of leukemic cells and pave the way to new studies aimed at testing GroPIns in combination therapies for the treatment of CLL.
Collapse
Affiliation(s)
| | - Alessia Varone
- Institute of Endocrinology and Experimental Oncology “G. Salvatore”, National Research Council, Naples, Italy
| | | | - Nagaja Capitani
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Federica Frezzato
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy
| | - Andrea Visentin
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy
| | - Livio Trentin
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padua, Italy
| | - Daniela Corda
- Department of Biomedical Sciences, National Research Council, Rome, Italy
| | | | - Laura Patrussi
- Department of Life Sciences, University of Siena, Siena, Italy
| |
Collapse
|
10
|
Kiratikanon S, Chattipakorn SC, Chattipakorn N, Kumfu S. The regulatory effects of PTPN6 on inflammatory process: Reports from mice to men. Arch Biochem Biophys 2022; 721:109189. [DOI: 10.1016/j.abb.2022.109189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 02/24/2022] [Accepted: 03/14/2022] [Indexed: 12/30/2022]
|
11
|
Tong JF, Zhou L, Li S, Lu LF, Li ZC, Li Z, Gan RH, Mou CY, Zhang QY, Wang ZW, Zhang XJ, Wang Y, Gui JF. Two Duplicated Ptpn6 Homeologs Cooperatively and Negatively Regulate RLR-Mediated IFN Response in Hexaploid Gibel Carp. Front Immunol 2021; 12:780667. [PMID: 34899743 PMCID: PMC8662705 DOI: 10.3389/fimmu.2021.780667] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/11/2021] [Indexed: 01/28/2023] Open
Abstract
Src homology region 2 domain-containing phosphatase 1 (SHP1), encoded by the protein tyrosine phosphatase nonreceptor type 6 (ptpn6) gene, belongs to the family of protein tyrosine phosphatases (PTPs) and participates in multiple signaling pathways of immune cells. However, the mechanism of SHP1 in regulating fish immunity is largely unknown. In this study, we first identified two gibel carp (Carassius gibelio) ptpn6 homeologs (Cgptpn6-A and Cgptpn6-B), each of which had three alleles with high identities. Then, relative to Cgptpn6-B, dominant expression in adult tissues and higher upregulated expression of Cgptpn6-A induced by polyinosinic-polycytidylic acid (poly I:C), poly deoxyadenylic-deoxythymidylic (dA:dT) acid and spring viremia of carp virus (SVCV) were uncovered. Finally, we demonstrated that CgSHP1-A (encoded by the Cgptpn6-A gene) and CgSHP1-B (encoded by the Cgptpn6-B gene) act as negative regulators of the RIG-I-like receptor (RLR)-mediated interferon (IFN) response via two mechanisms: the inhibition of CaTBK1-induced phosphorylation of CaMITA shared by CgSHP1-A and CgSHP1-B, and the autophagic degradation of CaMITA exclusively by CgSHP1-A. Meanwhile, the data support that CgSHP1-A and CgSHP1-B have sub-functionalized and that CgSHP1-A overwhelmingly dominates CgSHP1-B in the process of RLR-mediated IFN response. The current study not only sheds light on the regulative mechanism of SHP1 in fish immunity, but also provides a typical case of duplicated gene evolutionary fates.
Collapse
Affiliation(s)
- Jin-Feng Tong
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovative Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.,Hubei Hongshan Laboratory, Chinese Academy of Sciences, Wuhan, China
| | - Li Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovative Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.,Hubei Hongshan Laboratory, Chinese Academy of Sciences, Wuhan, China
| | - Shun Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovative Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Long-Feng Lu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovative Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Zhuo-Cong Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovative Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Zhi Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovative Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China.,Hubei Hongshan Laboratory, Chinese Academy of Sciences, Wuhan, China
| | - Rui-Hai Gan
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovative Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.,Hubei Hongshan Laboratory, Chinese Academy of Sciences, Wuhan, China
| | - Cheng-Yan Mou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovative Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.,Fisheries Institute, Sichuan Academy of Agricultural Sciences, Chengdu, China
| | - Qi-Ya Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovative Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Zhong-Wei Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovative Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.,Hubei Hongshan Laboratory, Chinese Academy of Sciences, Wuhan, China
| | - Xiao-Juan Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovative Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China
| | - Yang Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovative Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.,Hubei Hongshan Laboratory, Chinese Academy of Sciences, Wuhan, China
| | - Jian-Fang Gui
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovative Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.,Hubei Hongshan Laboratory, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
12
|
Hao F, Wang C, Sholy C, Cao M, Kang X. Strategy for Leukemia Treatment Targeting SHP-1,2 and SHIP. Front Cell Dev Biol 2021; 9:730400. [PMID: 34490276 PMCID: PMC8417302 DOI: 10.3389/fcell.2021.730400] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 07/28/2021] [Indexed: 11/29/2022] Open
Abstract
Protein tyrosine phosphatases (PTPs) are modulators of cellular functions such as differentiation, metabolism, migration, and survival. PTPs antagonize tyrosine kinases by removing phosphate moieties from molecular signaling residues, thus inhibiting signal transduction. Two PTPs, SHP-1 and SHP-2 (SH2 domain-containing phosphatases 1 and 2, respectively) and another inhibitory phosphatase, SH2 domain-containing inositol phosphatase (SHIP), are essential for cell function, which is reflected in the defective phenotype of mutant mice. Interestingly, SHP-1, SHP-2, and SHIP mutations are identified in many cases of human leukemia. However, the impact of these phosphatases and their mutations regarding the onset and progression of leukemia is controversial. The ambiguity of the role of these phosphatases imposes challenges on the development of targeting therapies for leukemia. This fundamental problem, confronted by the expanding investigational field of leukemia, will be addressed in this review, which will include a discussion of the molecular mechanisms of SHP-1, SHP-2, and SHIP in normal hematopoiesis and their role in leukemia. Clinical development of leukemic therapies achieved by targeting these phosphatases will be addressed as well.
Collapse
Affiliation(s)
- Fang Hao
- Center for Precision Medicine, Department of Medicine, University of Missouri, Columbia, MO, United States
| | - Chen Wang
- Center for Precision Medicine, Department of Medicine, University of Missouri, Columbia, MO, United States
| | - Christine Sholy
- Center for Precision Medicine, Department of Medicine, University of Missouri, Columbia, MO, United States
| | - Min Cao
- Center for Precision Medicine, Department of Medicine, University of Missouri, Columbia, MO, United States
| | - Xunlei Kang
- Center for Precision Medicine, Department of Medicine, University of Missouri, Columbia, MO, United States
| |
Collapse
|
13
|
Chen L, Bao Y, Jiang S, Zhong XB. The Roles of Long Noncoding RNAs HNF1α-AS1 and HNF4α-AS1 in Drug Metabolism and Human Diseases. Noncoding RNA 2020; 6:E24. [PMID: 32599764 PMCID: PMC7345002 DOI: 10.3390/ncrna6020024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/15/2020] [Accepted: 06/22/2020] [Indexed: 12/12/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) are RNAs with a length of over 200 nucleotides that do not have protein-coding abilities. Recent studies suggest that lncRNAs are highly involved in physiological functions and diseases. lncRNAs HNF1α-AS1 and HNF4α-AS1 are transcripts of lncRNA genes HNF1α-AS1 and HNF4α-AS1, which are antisense lncRNA genes located in the neighborhood regions of the transcription factor (TF) genes HNF1α and HNF4α, respectively. HNF1α-AS1 and HNF4α-AS1 have been reported to be involved in several important functions in human physiological activities and diseases. In the liver, HNF1α-AS1 and HNF4α-AS1 regulate the expression and function of several drug-metabolizing cytochrome P450 (P450) enzymes, which also further impact P450-mediated drug metabolism and drug toxicity. In addition, HNF1α-AS1 and HNF4α-AS1 also play important roles in the tumorigenesis, progression, invasion, and treatment outcome of several cancers. Through interacting with different molecules, including miRNAs and proteins, HNF1α-AS1 and HNF4α-AS1 can regulate their target genes in several different mechanisms including miRNA sponge, decoy, or scaffold. The purpose of the current review is to summarize the identified functions and mechanisms of HNF1α-AS1 and HNF4α-AS1 and to discuss the future directions of research of these two lncRNAs.
Collapse
Affiliation(s)
- Liming Chen
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT 06269, USA; (L.C.); (Y.B.); (S.J.)
| | - Yifan Bao
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT 06269, USA; (L.C.); (Y.B.); (S.J.)
| | - Suzhen Jiang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT 06269, USA; (L.C.); (Y.B.); (S.J.)
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 51006, China
| | - Xiao-bo Zhong
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT 06269, USA; (L.C.); (Y.B.); (S.J.)
| |
Collapse
|
14
|
Varone A, Spano D, Corda D. Shp1 in Solid Cancers and Their Therapy. Front Oncol 2020; 10:935. [PMID: 32596156 PMCID: PMC7300250 DOI: 10.3389/fonc.2020.00935] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 05/12/2020] [Indexed: 12/20/2022] Open
Abstract
Shp1 is a cytosolic tyrosine phosphatase that regulates a broad range of cellular functions and targets, modulating the flow of information from the cell membrane to the nucleus. While initially studied in the hematopoietic system, research conducted over the past years has expanded our understanding of the biological role of Shp1 to other tissues, proposing it as a novel tumor suppressor gene functionally involved in different hallmarks of cancer. The main mechanism by which Shp1 curbs cancer development and progression is the ability to attenuate and/or terminate signaling pathways controlling cell proliferation, survival, migration, and invasion. Thus, alterations in Shp1 function or expression can contribute to several human diseases, particularly cancer. In cancer cells, Shp1 activity can indeed be affected by mutations or epigenetic silencing that cause failure of Shp1-mediated homeostatic maintenance. This review will discuss the current knowledge of the cellular functions controlled by Shp1 in non-hematopoietic tissues and solid tumors, the mechanisms that regulate Shp1 expression, the role of its mutation/expression status in cancer and its value as potential target for cancer treatment. In addition, we report information gathered from the public available data from The Cancer Genome Atlas (TCGA) database on Shp1 genomic alterations and correlation with survival in solid cancers patients.
Collapse
Affiliation(s)
- Alessia Varone
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Daniela Spano
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Daniela Corda
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy.,Department of Biomedical Sciences, National Research Council, Rome, Italy
| |
Collapse
|
15
|
Wang Y, Wang P, Xu J. Phosphorylation: A Fast Switch For Checkpoint Signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1248:347-398. [PMID: 32185718 DOI: 10.1007/978-981-15-3266-5_15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Checkpoint signaling involves a variety of upstream and downstream factors that participate in the regulation of checkpoint expression, activation, and degradation. During the process, phosphorylation plays a critical role. Phosphorylation is one of the most well-documented post-translational modifications of proteins. Of note, the importance of phosphorylation has been emphasized in aspects of cell activities, including proliferation, metabolism, and differentiation. Here we summarize how phosphorylation of specific molecules affects the immune activities with preference in tumor immunity. Of course, immune checkpoints are given extra attention in this book. There are many common pathways that are involved in signaling of different checkpoints. Some of them are integrated and presented as common activities in the early part of this chapter, especially those associated with PD-1/PD-L1 and CTLA-4, because investigations concerning them are particularly abundant and variant. Their distinct regulation is supplementarily discussed in their respective section. As for checkpoints that are so far not well explored, their related phosphorylation modulations are listed separately in the later part. We hope to provide a clear and systematic view of the phosphorylation-modulated immune signaling.
Collapse
Affiliation(s)
- Yiting Wang
- School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ping Wang
- Shanghai Tenth People's Hospital of Tongji University, School of Medicine, School of Life Sciences and Technology, Tongji University Cancer Center, Tongji University, Shanghai, 200092, China
| | - Jie Xu
- Institutes of Biomedical Sciences, Zhongshan-Xuhui Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
16
|
Tian H, Tan R, Ye B, Yan S, Sui M, Zhao W, Zhang L, Zhu Y, Zeng L. SHP-1 inhibits renal ischemia reperfusion injury via dephosphorylating ASK1 and suppressing apoptosis. Biochem Biophys Res Commun 2019; 513:360-367. [PMID: 30961932 DOI: 10.1016/j.bbrc.2019.03.187] [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: 03/18/2019] [Accepted: 03/27/2019] [Indexed: 12/14/2022]
Abstract
Apoptosis of tubular epithelium cells (TECs) plays critical roles in renal ischemia reperfusion (I/R) injury, but the molecular regulatory mechanisms of apoptosis still require further investigation. Recently, phosphatase family members have been suggested to regulate multiple aspects of the injury and regeneration response. However, the roles of SHP-1, an important protein-tyrosine phosphatase, in the regulation of renal I/R injury remain unknown. Here, we found that SHP-1 knockdown in vivo significantly increased renal I/R injury and aggravated the apoptosis of TECs. Consistently, after SHP-1 knockdown in TECs in vitro, a sharp increase of apoptosis induced by cobalt dichloride was found. The protective role of SHP-1 was also validated in a TEC cell line stably overexpressing SHP-1. Mechanistically, the ASK1/MKK4/JNK pro-apoptosis signal was over activated after SHP-1 knockdown, and SHP-1 could bind to and dephosphorylate ASK1 to inhibit its activation, thus repressing apoptosis.
Collapse
Affiliation(s)
- Hongzhe Tian
- Department of Organ Transplantation, Changhai Hospital, Second Military Medical University, Shanghai, 200433, PR China
| | - Rumeng Tan
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Bogen Ye
- Department of Organ Transplantation, Changhai Hospital, Second Military Medical University, Shanghai, 200433, PR China
| | - Sijia Yan
- Department of Organ Transplantation, Changhai Hospital, Second Military Medical University, Shanghai, 200433, PR China
| | - Mingxing Sui
- Department of Organ Transplantation, Changhai Hospital, Second Military Medical University, Shanghai, 200433, PR China
| | - Wenyu Zhao
- Department of Organ Transplantation, Changhai Hospital, Second Military Medical University, Shanghai, 200433, PR China
| | - Lei Zhang
- Department of Organ Transplantation, Changhai Hospital, Second Military Medical University, Shanghai, 200433, PR China
| | - Youhua Zhu
- Department of Organ Transplantation, Changhai Hospital, Second Military Medical University, Shanghai, 200433, PR China
| | - Li Zeng
- Department of Organ Transplantation, Changhai Hospital, Second Military Medical University, Shanghai, 200433, PR China.
| |
Collapse
|
17
|
Wang H, Morris RG, Knepper MA, Zhou X. Sickle cell disease up-regulates vasopressin, aquaporin 2, urea transporter A1, Na-K-Cl cotransporter 2, and epithelial Na channels in the mouse kidney medulla despite compromising urinary concentration ability. Physiol Rep 2019; 7:e14066. [PMID: 31033226 PMCID: PMC6487471 DOI: 10.14814/phy2.14066] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 03/18/2019] [Accepted: 03/29/2019] [Indexed: 11/24/2022] Open
Abstract
Sickle cell disease (SCD)-induced urinary concentration defect has been proposed as caused by impaired ability of the occluded vasa recta due to red blood cell sickling to serve as countercurrent exchangers and renal tubules to absorb water and solutes. However, the exact molecular mechanisms remain largely unknown. The present studies were undertaken to determine the effects of SCD on vasopressin, aquaporin2 (AQP2), urea transporter A1 (UTA1), Na-K-Cl co-transporter 2 (NKCC2), epithelial Na channels (ENaC), aquaporin1 (AQP1), nuclear factor of activated T cells 5 (NFAT5) and Src homology region-2 domain-containing phosphatase-1 (SHP-1), an important regulator of NFAT5, in the Berkeley SCD mouse kidney medulla. Under water repletion, SCD only induced a minor urinary concentration defect associated with increased urinary vasopressin level alone with the well-known effects of vasopressin: protein abundance of AQP2, UTA1 and ENaC-β and apical targeting of AQP2 as compared with non-SCD. SCD did not significantly affect AQP1 protein level. Water restriction had no further significant effect on SCD urinary vasopressin. NFAT5 is also critical to urinary concentration. Instead, water restriction-activated NFAT5 associated with inhibition of SHP-1 in the SCD mice. Yet, water restriction only elevated urinary osmolality by 28% in these mice as opposed to 104% in non-SCD mice despite similar degree increases of protein abundance of AQP2, NKCC2 and AQP2-S256-P. Water-restriction had no significant effect on protein abundance of ENaC or AQP1 in either strain. In conclusion, under water repletion SCD, only induces a minor defect in urinary concentration because of compensation from the up-regulated vasopressin system. However, under water restriction, SCD mice struggle to concentrate urine despite activating NFAT5. SCD-induced urinary concentration defect appears to be resulted from the poor blood flow in vasa recta rather than the renal tubules' ability to absorb water and solutes.
Collapse
Affiliation(s)
- Hong Wang
- Department of MedicineUniformed Services University of Health SciencesBethesdaMaryland
| | | | | | - Xiaoming Zhou
- Department of MedicineUniformed Services University of Health SciencesBethesdaMaryland
| |
Collapse
|
18
|
Liu L, Zhang S, Liu X, Liu J. Aberrant promoter 2 methylation‑mediated downregulation of protein tyrosine phosphatase, non‑receptor type 6, is associated with progression of esophageal squamous cell carcinoma. Mol Med Rep 2019; 19:3273-3282. [PMID: 30816454 DOI: 10.3892/mmr.2019.9971] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 02/05/2019] [Indexed: 11/05/2022] Open
Abstract
The human protein tyrosine phosphatase, non‑receptor type 6 (PTPN6) gene is located on chromosome 12p13 and encodes an Mr 68,000 non‑receptor type protein‑tyrosine phosphatase. The PTPN6 gene has been considered as a candidate tumor suppressor in hematological and solid malignancies, and promoter methylation may be an epigenetic modification silencing its expression. However, the detailed role of PTPN6 and its promoter methylation status in the pathogenesis of esophageal squamous cell carcinoma (ESCC) has not been fully elucidated. The aim of the present study was to investigate PTPN6 expression in ESCC tissues and esophageal cancer cell lines, detect the effect of CpG hypermethylation on the activity of PTPN6, and additionally elucidate the role and prognostic significance of PTPN6 in ESCC tumorigenesis and progression. The expression of PTPN6 was identified to be significantly downregulated in esophageal cancer cell lines and ESCC tissues. Marked upregulation of PTPN6 was detected in 5‑aza‑2'‑deoxycytidine‑treated esophageal cancer cells, and frequent hypermethylation of the CpG sites within the P2 promoter (P2) was detected in ESCC tissues and esophageal cancer cell lines. The expression and methylation status of PTPN6 was associated with tumor node metastasis stage, pathological differentiation and lymph node metastasis in patients with ESCC. Aberrant hypermethylation of the P2 exhibited marked tumor specificity and was identified to be associated with the expression level of PTPN6. Downregulation and hypermethylation of PTPN6 were identified to be associated with poor ESCC patient survival. Furthermore, upregulation of PTPN6 inhibited the proliferation and invasion of esophageal cancer cells in vitro. The results of the present study suggest that PTPN6 may serve as a tumor suppressor in ESCC, and it may serve as a potential target for antitumor therapy.
Collapse
Affiliation(s)
- Lei Liu
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Shaowei Zhang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Xinbo Liu
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Junfeng Liu
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
19
|
Jiang N, Lin JJ, Wang J, Zhang BN, Li A, Chen ZY, Guo S, Li BB, Duan YZ, Yan RY, Yan HF, Fu XY, Zhou JL, Yang HM, Cui Y. Novel treatment strategies for patients with HER2-positive breast cancer who do not benefit from current targeted therapy drugs. Exp Ther Med 2018; 16:2183-2192. [PMID: 30186457 PMCID: PMC6122384 DOI: 10.3892/etm.2018.6459] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 06/27/2018] [Indexed: 12/11/2022] Open
Abstract
Human epidermal growth factor receptor-2 positive breast cancer (HER2+ BC) is characterized by a high rate of metastasis and drug resistance. The advent of targeted therapy drugs greatly improves the prognosis of HER2+ BC patients. However, drug resistance or severe side effects have limited the application of targeted therapy drugs. To achieve more effective treatment, considerable research has concentrated on strategies to overcome drug resistance. Abemaciclib (CDK4/6 inhibitor), a new antibody-drug conjugate (ADC), src homology 2 (SH2) containing tyrosine phosphatase-1 (SHP-1) and fatty acid synthase (FASN) have been demonstrated to improve drug resistance. In addition, using an effective vector to accurately deliver drugs to tumors has shown good application prospects. Many studies have also found that natural anti-cancer substances produced effective results during in vitro and in vivo anti-HER2+ BC research. This review aimed to summarize the current status of potential clinical drugs that may benefit HER2+ BC patients in the future.
Collapse
Affiliation(s)
- Nan Jiang
- Department of General Surgery, 306 Hospital of PLA, Beijing 100101, P.R. China
- Department of General Surgery, 306 Teaching Hospital of Peking University Health Science Center, Beijing 100101, P.R. China
| | - Jing-Jing Lin
- Department of General Surgery, 306 Hospital of PLA, Beijing 100101, P.R. China
- Department of General Surgery, 306 Teaching Hospital of Peking University Health Science Center, Beijing 100101, P.R. China
| | - Jun Wang
- Department of Hepatology, 302 Teaching Hospital of Peking University Health Science Center, Beijing 100101, P.R. China
| | - Bei-Ning Zhang
- Department of General Surgery, 306 Hospital of PLA, Beijing 100101, P.R. China
- Department of General Surgery, PLA 306 Clinical Hospital of Anhui Medical University, Beijing 230000, P.R. China
| | - Ao Li
- Department of General Surgery, 306 Hospital of PLA, Beijing 100101, P.R. China
- Department of General Surgery, PLA 306 Clinical Hospital of Anhui Medical University, Beijing 230000, P.R. China
| | - Zheng-Yang Chen
- Department of General Surgery, 306 Hospital of PLA, Beijing 100101, P.R. China
- Department of General Surgery, 306 Teaching Hospital of Peking University Health Science Center, Beijing 100101, P.R. China
| | - Song Guo
- Department of General Surgery, 306 Hospital of PLA, Beijing 100101, P.R. China
- Department of General Surgery, 306 Teaching Hospital of Peking University Health Science Center, Beijing 100101, P.R. China
| | - Bin-Bin Li
- Department of General Surgery, 306 Hospital of PLA, Beijing 100101, P.R. China
- Department of General Surgery, PLA 306 Clinical Hospital of Anhui Medical University, Beijing 230000, P.R. China
| | - Yu-Zhong Duan
- Department of General Surgery, 306 Hospital of PLA, Beijing 100101, P.R. China
| | - Ru-Yi Yan
- Department of General Surgery, PLA 306 Clinical Hospital of Anhui Medical University, Beijing 230000, P.R. China
- Department of Pathology, 306 Hospital of PLA, Beijing 100101, P.R. China
| | - Hong-Feng Yan
- Department of General Surgery, 306 Hospital of PLA, Beijing 100101, P.R. China
| | - Xiao-Yan Fu
- Department of General Surgery, 306 Hospital of PLA, Beijing 100101, P.R. China
| | - Jin-Lian Zhou
- Department of Pathology, 306 Hospital of PLA, Beijing 100101, P.R. China
| | - He-Ming Yang
- Department of General Surgery, 306 Hospital of PLA, Beijing 100101, P.R. China
| | - Yan Cui
- Department of General Surgery, 306 Hospital of PLA, Beijing 100101, P.R. China
| |
Collapse
|
20
|
Ding CH, Yin C, Chen SJ, Wen LZ, Ding K, Lei SJ, Liu JP, Wang J, Chen KX, Jiang HL, Zhang X, Luo C, Xie WF. The HNF1α-regulated lncRNA HNF1A-AS1 reverses the malignancy of hepatocellular carcinoma by enhancing the phosphatase activity of SHP-1. Mol Cancer 2018; 17:63. [PMID: 29466992 PMCID: PMC5822613 DOI: 10.1186/s12943-018-0813-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 02/08/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Our previous study has demonstrated that hepatocyte nuclear factor 1α (HNF1α) exerts potent therapeutic effects on hepatocellular carcinoma (HCC). However, the molecular mechanisms by which HNF1α reverses HCC malignancy need to be further elucidated. METHODS lncRNA microarray was performed to identify the long noncoding RNAs (lncRNAs) regulated by HNF1α. Chromatin immunoprecipitation and luciferase reporter assays were applied to clarify the mechanism of the transcriptional regulation of HNF1α to HNF1A antisense RNA 1 (HNF1A-AS1). The effect of HNF1A-AS1 on HCC malignancy was evaluated in vitro and in vivo. RNA pulldown, RNA-binding protein immunoprecipitation and the Bio-Layer Interferometry assay were used to validate the interaction of HNF1A-AS1 and Src homology region 2 domain-containing phosphatase 1 (SHP-1). RESULTS HNF1α regulated the expression of a subset of lncRNAs in HCC cells. Among these lncRNAs, the expression levels of HNF1A-AS1 were notably correlated with HNF1α levels in HCC cells and human HCC tissues. HNF1α activated the transcription of HNF1A-AS1 by directly binding to its promoter region. HNF1A-AS1 inhibited the growth and the metastasis of HCC cells in vitro and in vivo. Moreover, knockdown of HNF1A-AS1 reversed the suppressive effects of HNF1α on the migration and invasion of HCC cells. Importantly, HNF1A-AS1 directly bound to the C-terminal of SHP-1 with a high binding affinity (KD = 59.57 ± 14.29 nM) and increased the phosphatase activity of SHP-1. Inhibition of SHP-1 enzymatic activity substantially reversed the HNF1α- or HNF1A-AS1-induced reduction on the metastatic property of HCC cells. CONCLUSIONS Our data revealed that HNF1A-AS1 is a direct transactivation target of HNF1α in HCC cells and involved in the anti-HCC effect of HNF1α. HNF1A-AS1 functions as phosphatase activator through the direct interaction with SHP-1. These findings suggest that regulation of the HNF1α/HNF1A-AS1/SHP-1 axis may have beneficial effects in the treatment of HCC.
Collapse
Affiliation(s)
- Chen-Hong Ding
- Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Chuan Yin
- Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Shi-Jie Chen
- Drug Discovery and Design Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai, 201203, China
| | - Liang-Zhi Wen
- Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, China.,Present address: Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Kai Ding
- Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Shu-Juan Lei
- Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Jin-Pei Liu
- Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Jian Wang
- Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Kai-Xian Chen
- Drug Discovery and Design Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai, 201203, China
| | - Hua-Liang Jiang
- Drug Discovery and Design Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai, 201203, China
| | - Xin Zhang
- Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Cheng Luo
- Drug Discovery and Design Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences (CAS), Shanghai, 201203, China.
| | - Wei-Fen Xie
- Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, China.
| |
Collapse
|
21
|
Dehkhoda F, Lee CMM, Medina J, Brooks AJ. The Growth Hormone Receptor: Mechanism of Receptor Activation, Cell Signaling, and Physiological Aspects. Front Endocrinol (Lausanne) 2018; 9:35. [PMID: 29487568 PMCID: PMC5816795 DOI: 10.3389/fendo.2018.00035] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 01/29/2018] [Indexed: 01/02/2023] Open
Abstract
The growth hormone receptor (GHR), although most well known for regulating growth, has many other important biological functions including regulating metabolism and controlling physiological processes related to the hepatobiliary, cardiovascular, renal, gastrointestinal, and reproductive systems. In addition, growth hormone signaling is an important regulator of aging and plays a significant role in cancer development. Growth hormone activates the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling pathway, and recent studies have provided a new understanding of the mechanism of JAK2 activation by growth hormone binding to its receptor. JAK2 activation is required for growth hormone-mediated activation of STAT1, STAT3, and STAT5, and the negative regulation of JAK-STAT signaling comprises an important step in the control of this signaling pathway. The GHR also activates the Src family kinase signaling pathway independent of JAK2. This review covers the molecular mechanisms of GHR activation and signal transduction as well as the physiological consequences of growth hormone signaling.
Collapse
Affiliation(s)
- Farhad Dehkhoda
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Christine M. M. Lee
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Johan Medina
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Andrew J. Brooks
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
22
|
Tibaldi E, Pagano MA, Frezzato F, Trimarco V, Facco M, Zagotto G, Ribaudo G, Pavan V, Bordin L, Visentin A, Zonta F, Semenzato G, Brunati AM, Trentin L. Targeted activation of the SHP-1/PP2A signaling axis elicits apoptosis of chronic lymphocytic leukemia cells. Haematologica 2017; 102:1401-1412. [PMID: 28619847 PMCID: PMC5541874 DOI: 10.3324/haematol.2016.155747] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 06/14/2017] [Indexed: 01/07/2023] Open
Abstract
Lyn, a member of the Src family of kinases, is a key factor in the dysregulation of survival and apoptotic pathways of malignant B cells in chronic lymphocytic leukemia. One of the effects of Lyn’s action is spatial and functional segregation of the tyrosine phosphatase SHP-1 into two pools, one beneath the plasma membrane in an active state promoting pro-survival signals, the other in the cytosol in an inhibited conformation and unable to counter the elevated level of cytosolic tyrosine phosphorylation. We herein show that SHP-1 activity can be elicited directly by nintedanib, an agent also known as a triple angiokinase inhibitor, circumventing the phospho-S591-dependent inhibition of the phosphatase, leading to the dephosphorylation of pro-apoptotic players such as procaspase-8 and serine/threonine phosphatase 2A, eventually triggering apoptosis. Furthermore, the activation of PP2A by using MP07-66, a novel FTY720 analog, stimulated SHP-1 activity via dephosphorylation of phospho-S591, which unveiled the existence of a positive feedback signaling loop involving the two phosphatases. In addition to providing further insights into the molecular basis of this disease, our findings indicate that the PP2A/SHP-1 axis may emerge as an attractive, novel target for the development of alternative strategies in the treatment of chronic lymphocytic leukemia.
Collapse
Affiliation(s)
- Elena Tibaldi
- Department of Molecular Medicine, University of Padua, Italy
| | - Mario Angelo Pagano
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Italy
| | - Federica Frezzato
- Department of Medicine, University of Padua, Italy.,Venetian Institute of Molecular Medicine (VIMM), Centro di Eccellenza per la Ricerca Biomedica, Padua, Italy
| | - Valentina Trimarco
- Department of Medicine, University of Padua, Italy.,Venetian Institute of Molecular Medicine (VIMM), Centro di Eccellenza per la Ricerca Biomedica, Padua, Italy
| | - Monica Facco
- Department of Medicine, University of Padua, Italy.,Venetian Institute of Molecular Medicine (VIMM), Centro di Eccellenza per la Ricerca Biomedica, Padua, Italy
| | - Giuseppe Zagotto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Italy
| | - Giovanni Ribaudo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Italy
| | - Valeria Pavan
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Italy
| | - Luciana Bordin
- Department of Molecular Medicine, University of Padua, Italy
| | - Andrea Visentin
- Department of Medicine, University of Padua, Italy.,Venetian Institute of Molecular Medicine (VIMM), Centro di Eccellenza per la Ricerca Biomedica, Padua, Italy
| | - Francesca Zonta
- Department of Biomedical Sciences, University of Padua, Italy
| | - Gianpietro Semenzato
- Department of Medicine, University of Padua, Italy.,Venetian Institute of Molecular Medicine (VIMM), Centro di Eccellenza per la Ricerca Biomedica, Padua, Italy
| | | | - Livio Trentin
- Department of Medicine, University of Padua, Italy.,Venetian Institute of Molecular Medicine (VIMM), Centro di Eccellenza per la Ricerca Biomedica, Padua, Italy
| |
Collapse
|
23
|
Demosthenous C, Han JJ, Hu G, Stenson M, Gupta M. Loss of function mutations in PTPN6 promote STAT3 deregulation via JAK3 kinase in diffuse large B-cell lymphoma. Oncotarget 2016; 6:44703-13. [PMID: 26565811 PMCID: PMC4792586 DOI: 10.18632/oncotarget.6300] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 10/22/2015] [Indexed: 11/25/2022] Open
Abstract
PTPN6 (SHP1) is a tyrosine phosphatase that negatively controls the activity of multiple signaling pathways including STAT signaling, however role of mutated PTPN6 is not much known. Here we investigated whether PTPN6 might also be a potential target for diffuse large B cell lymphoma (DLBCL) and performed Sanger sequencing of the PTPN6 gene. We have identified missense mutations within PTPN6 (N225K and A550V) in 5% (2/38) of DLBCL tumors. Site directed mutagenesis was performed to mutate wild type (WT) PTPN6 and stable cell lines were generated by lentiviral transduction of PTPN6(WT), PTPN6(N225K) and PTPN6(A550V) constructs, and effects of WT or mutated PTPN6 on STAT3 signaling were analyzed. WT PTPN6 dephosphorylated STAT3, but had no effect on STAT1, STAT5 or STAT6 phosphorylation. Both PTPN6 mutants were unable to inhibit constitutive, as well as cytokines induced STAT3 activation. Both PTPN6 mutants also demonstrated reduced tyrosine phosphatase activity and exhibited enhanced STAT3 transactivation activity. Intriguingly, a lack of direct binding between STAT3 and WT or mutated PTPN6 was observed. However, compared to WT PTPN6, cells expressing PTPN6 mutants exhibited increased binding between JAK3 and PTPN6 suggesting a more dynamic interaction of PTPN6 with upstream regulators of STAT3. Consistent with this notion, both the mutants demonstrated increased resistance to JAK3 inhibitor, WHIP-154 relative to WT PTPN6. Overall, this is the first study, which demonstrates that N225K and A550V PTPN6 mutations cause loss-of-function leading to JAK3 mediated deregulation of STAT3 pathway and uncovers a mechanism that tumor cells can use to control PTPN6 substrate specificity.
Collapse
Affiliation(s)
- Christos Demosthenous
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - Jing Jing Han
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - Guangzhen Hu
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - Mary Stenson
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - Mamta Gupta
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, College of Medicine, Rochester, MN, USA
| |
Collapse
|
24
|
Abstract
The neutrophil transmigration across the blood endothelial cell barrier represents the prerequisite step of innate inflammation. Neutrophil recruitment to inflamed tissues occurs in a well-defined stepwise manner, which includes elements of neutrophil rolling, firm adhesion, and crawling onto the endothelial cell surface before transmigrating across the endothelial barrier. This latter step known as diapedesis can occur at the endothelial cell junction (paracellular) or directly through the endothelial cell body (transcellular). The extravasation cascade is controlled by series of engagement of various adhesive modules, which result in activation of bidirectional signals to neutrophils and endothelial cells for adequate cellular response. This review will focus on recent advances in our understanding of mechanism of leukocyte crawling and diapedesis, with an emphasis on leukocyte-endothelial interactions and the signaling pathways they transduce to determine the mode of diapedesis, junctional or nonjunctional. I will also discuss emerging evidence highlighting key differences in the two modes of diapedesis and why it is clinically important to understand specificity in the regulation of diapedesis.
Collapse
Affiliation(s)
- Marie-Dominique Filippi
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Research Foundation, Cincinnati, Ohio, USA; University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.
| |
Collapse
|
25
|
Wu Y, Li R, Zhang J, Wang G, Liu B, Huang X, Zhang T, Luo R. Protein tyrosine phosphatase SHP-1 sensitizes EGFR/HER-2 positive breast cancer cells to trastuzumab through modulating phosphorylation of EGFR and HER-2. Onco Targets Ther 2015; 8:2577-87. [PMID: 26396531 PMCID: PMC4576899 DOI: 10.2147/ott.s82225] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Trastuzumab resistance in HER-2 positive breast cancer cells is closely related to overexpression of both epidermal growth factor receptor (EGFR) and human epidermal receptor (HER-2). SHP-1 has been demonstrated to downregulate tyrosine kinase activity including EGFR via its phosphatase function, but its effect on HER-2 activity is still unknown. Here, we examined the hypothesis that SHP-1 enhances the anticancer efficacy of trastuzumab in EGFR/HER-2 positive breast cancer cells through combining dual inhibition of EGFR and HER-2. Methods Trastuzumab-resistant breast cancer SKBr-3 cells were generated by long-term in vitro culture of SKBr-3cells in the presence of trastuzumab. The SHP-1 was ectopically expressed by stable transfection. The activity and expression of EGFR, HER-2, and downstream signaling pathways were tested by Western blot. Cell viability was examined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, and apoptosis was examined by flow cytometry. The binding between SHP-1 and EGFR/HER-2 was evaluated by immunoprecipitation assay and bimolecular fluorescence complementation. The effects of SHP-1 on tumorigenicity and trastuzumab sensitivity were confirmed via in vivo xenograft model. Results Trastuzumab-resistant SKBr-3 cells showed aberrant co-expression of EGFR and HER-2. Introduction of wild-type SHP-1 inhibited cell proliferation, clone formation, and promoted the apoptosis induced by trastuzumab. Meanwhile, SHP-1 overexpression reduced phosphorylation levels of EGFR and HER-2 both in parental and trastuzumab-resistant SKBr-3 cells. In vivo study showed an increased antitumor effect of trastuzumab in SHP-1 overexpressed xenografts. At last, we discovered that SHP-1 can make complexes with both EGFR and HER-2, and both phospho-EGFR and phosphor-HER-2 levels in wild-type SHP-1 immunoprecipitates were less than those in phosphatase-inactive SHP-1 (C453S) immunoprecipitates, indicating that EGFR and HER-2 are potential substrates of SHP-1. Conclusion Taken together, we have demonstrated that the SHP-1 is a negative regulatory factor of the tyrosine kinase activity of HER-2 and EGFR through inhibiting phosphorylation. Dual targeting of EGFR and HER-2, by combining trastuzumab with SHP-1 overexpression, may improve response in HER-2 overexpressing breast cancer cells that also express high levels of EGFR.
Collapse
Affiliation(s)
- Yifen Wu
- Traditional Chinese Medicine-Integrated Hospital, Southern Medical University, Guangzhou, People's Republic of China ; Department of Oncology, Dongguan People's Hospital, Dongguan, People's Republic of China
| | - Rong Li
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Junyi Zhang
- Department of Oncology, Traditional Chinese Medicine-Integrated Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Gang Wang
- Department of Radiology, Dongguan People's Hospital, Dongguan, People's Republic of China
| | - Bin Liu
- Second Affiliated Hospital of Guangzhou Medical College, Southern Medical University, Guangdong, People's Republic of China
| | - Xiaofang Huang
- College of Traditional Chinese medicine, Southern Medical University, Guangdong, People's Republic of China
| | - Tao Zhang
- College of Traditional Chinese medicine, Southern Medical University, Guangdong, People's Republic of China
| | - Rongcheng Luo
- Cancer Center, Traditional Chinese Medicine-Integrated Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
26
|
|
27
|
An HNF1α-regulated feedback circuit modulates hepatic fibrogenesis via the crosstalk between hepatocytes and hepatic stellate cells. Cell Res 2015; 25:930-45. [PMID: 26169608 PMCID: PMC4528057 DOI: 10.1038/cr.2015.84] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Revised: 01/27/2015] [Accepted: 06/02/2015] [Indexed: 12/29/2022] Open
Abstract
Hepatocytes are critical for the maintenance of liver homeostasis, but its involvement in hepatic fibrogenesis remains elusive. Hepatocyte nuclear factor 1α (HNF1α) is a liver-enriched transcription factor that plays a key role in hepatocyte function. Our previous study revealed a significant inhibitory effect of HNF1α on hepatocellular carcinoma. In this study, we report that the expression of HNF1α is significantly repressed in both human and rat fibrotic liver. Knockdown of HNF1α in the liver significantly aggravates hepatic fibrogenesis in either dimethylnitrosamine (DMN) or bile duct ligation (BDL) model in rats. In contrast, forced expression of HNF1α markedly alleviates hepatic fibrosis. HNF1α regulates the transcriptional expression of SH2 domain-containing phosphatase-1 (SHP-1) via directly binding to SHP-1 promoter in hepatocytes. Inhibition of SHP-1 expression abrogates the anti-fibrotic effect of HNF1α in DMN-treated rats. Moreover, HNF1α repression in primary hepatocytes leads to the activation of NF-κB and JAK/STAT pathways and initiates an inflammatory feedback circuit consisting of HNF1α, SHP-1, STAT3, p65, miR-21 and miR-146a, which sustains the deregulation of HNF1α in hepatocytes. More interestingly, a coordinated crosstalk between hepatocytes and hepatic stellate cells (HSCs) participates in this positive feedback circuit and facilitates the progression of hepatocellular damage. Our findings demonstrate that impaired hepatocytes play an active role in hepatic fibrogenesis. Early intervention of HNF1α-regulated inflammatory feedback loop in hepatocytes may have beneficial effects in the treatment of chronic liver diseases.
Collapse
|
28
|
Stadtmann A, Block H, Volmering S, Abram C, Sohlbach C, Boras M, Lowell CA, Zarbock A. Cross-Talk between Shp1 and PIPKIγ Controls Leukocyte Recruitment. THE JOURNAL OF IMMUNOLOGY 2015; 195:1152-61. [PMID: 26101325 DOI: 10.4049/jimmunol.1500606] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 05/22/2015] [Indexed: 12/13/2022]
Abstract
Neutrophil recruitment to the site of inflammation plays a pivotal role in host defense. However, overwhelming activation and accumulation of neutrophils in the tissue may cause tissue damage and autoimmunity due to the release of cytokines, oxidants, and proteases. Neutrophil adhesion in acute inflammation is initiated by activation of αLβ2 (LFA-1), which can be induced by rolling on E-selectin (slowly) or by exposure to the chemokine CXCL1 (rapidly). Despite the clinical importance, cell-intrinsic molecular mechanisms of negative regulation of integrin adhesiveness and neutrophil recruitment are poorly understood. Mice deficient in the tyrosine phosphatase Src homology 2 domain-containing protein tyrosine phosphatase 1 (Shp1) show increased leukocyte adhesion, but the interpretation of these data is limited by the severe global phenotype of these mice. In this study, we used mice with global and myeloid-restricted deletion of Shp1 to study neutrophil arrest, adhesion, crawling, and transendothelial migration in vitro and in vivo. Shp1 deficiency results in increased neutrophil adhesion in vivo; however, neutrophil crawling, transmigration, and chemotaxis were reduced in these mice. Mechanistically, Shp1 binds and controls PIPKIγ activity and, thereby, modulates phosphatidylinositol (4,5)-bisphosphate levels and adhesion. Thus, Shp1 is involved in the deactivation of integrins and regulation of neutrophil recruitment into inflamed tissue.
Collapse
Affiliation(s)
- Anika Stadtmann
- Department of Anesthesiology, Intensive Care, and Pain Medicine, University of Münster, 48149 Münster, Germany; Max-Planck Institute for Molecular Biomedicine, 48149 Münster, Germany; and
| | - Helena Block
- Department of Anesthesiology, Intensive Care, and Pain Medicine, University of Münster, 48149 Münster, Germany; Max-Planck Institute for Molecular Biomedicine, 48149 Münster, Germany; and
| | - Stephanie Volmering
- Department of Anesthesiology, Intensive Care, and Pain Medicine, University of Münster, 48149 Münster, Germany; Max-Planck Institute for Molecular Biomedicine, 48149 Münster, Germany; and
| | - Clare Abram
- Department of Laboratory Medicine, University of California, San Francisco, CA 94143
| | - Charlotte Sohlbach
- Department of Anesthesiology, Intensive Care, and Pain Medicine, University of Münster, 48149 Münster, Germany; Max-Planck Institute for Molecular Biomedicine, 48149 Münster, Germany; and
| | - Mark Boras
- Department of Anesthesiology, Intensive Care, and Pain Medicine, University of Münster, 48149 Münster, Germany; Max-Planck Institute for Molecular Biomedicine, 48149 Münster, Germany; and
| | - Clifford A Lowell
- Department of Laboratory Medicine, University of California, San Francisco, CA 94143
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care, and Pain Medicine, University of Münster, 48149 Münster, Germany; Max-Planck Institute for Molecular Biomedicine, 48149 Münster, Germany; and
| |
Collapse
|
29
|
Filippi MD. Leukocyte transcellular diapedesis: Rap1b is in control. Tissue Barriers 2015; 3:e1052185. [PMID: 26451346 DOI: 10.1080/21688370.2015.1052185] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 05/11/2015] [Accepted: 05/13/2015] [Indexed: 12/30/2022] Open
Abstract
The neutrophil transmigration across the blood endothelial cell barrier represents the prerequisite step of innate inflammation. It is well known that neutrophils cross the endothelial barrier by transmigrating at the endothelial cell junction ('paracellular'). However, in vivo and in vitro evidence have clearly demonstrated occurrence of an alternate mode of migration directly through the endothelial cell body ('transcellular'). Despite our knowledge on mechanisms of transendothelial migration, it remains unclear which factors determine distinct modes of migration. We recently found that the Ras-like Rap1b GTPase limits neutrophil transcellular migration. Rap1b restrains transcellular migration by suppressing Akt-driven invasive protrusions while leaving the paracellular route unaffected. Furthermore, Rap1b limits neutrophil tissue infiltration in mice and prevents hyper susceptibility to endotoxin shock. These findings uncover a novel role for Rap1b in neutrophil migration and inflammation. Importantly, they offer emerging evidences that paracellular and transcellular migration of neutrophils are regulated by separate mechanisms. Here, we discuss the mechanisms of neutrophil transmigration and their clinical importance for vascular integrity and innate inflammation.
Collapse
Affiliation(s)
- Marie-Dominique Filippi
- Division of Experimental Hematology and Cancer Biology; Cincinnati Children's Research Foundation ; Cincinnati, OH USA ; University of Cincinnati College of Medicine ; Cincinnati, OH USA
| |
Collapse
|
30
|
Khan TH, Srivastava N, Srivastava A, Sareen A, Mathur RK, Chande AG, Musti KV, Roy S, Mukhopadhyaya R, Saha B. SHP-1 Plays a Crucial Role in CD40 Signaling Reciprocity. THE JOURNAL OF IMMUNOLOGY 2014; 193:3644-53. [DOI: 10.4049/jimmunol.1400620] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
31
|
Prostate anatomy in motheaten viable (me(v)) mice with mutations in the protein tyrosine phosphatase SHP-1. Actas Urol Esp 2014; 38:438-44. [PMID: 24819344 DOI: 10.1016/j.acuro.2014.02.005] [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: 02/03/2014] [Accepted: 02/06/2014] [Indexed: 11/23/2022]
Abstract
OBJECTIVE To study prostate and seminal vesicle anatomy in viable motheaten (mev) with mutations in PTPN6 gene leading to a severe reduction in the activity of protein tyrosine phosphatase SHP-1. Homozygous mev mice exhibit multiple anomalies that include immunodeficiencies, increased proliferation of macrophage, neutrophil, and erythrocyte progenitors, decreased bone density and sterility. MATERIAL AND METHOD We analyzed macro- and microscopic anatomy of the seminal vesicle and prostate macro- and microscopic anatomy of 5 mev/mev and 8 wt/wt adult 7 week old mice. Computerized morphometric analysis was performed to measure the relative changes appearing in the epithelial volume of the different prostatic lobes. RESULTS All mice studied revealed normal genital organs (penis, testis, epididymis, vas deferens) and bladder. The seminal vesicle was absent in all mev/mev individuals analyzed, being normal and very noticeable in wt/wt mice. The different glands that compose the prostatic complex (anterior, ventral and dorso-lateral prostate) were atrophied in mev/mev mice: anterior prostate 0.4 times, ventral 0.19 times, dorsal 0.35 times and lateral 0.28 times those of the respective regions in wt/wt mice. Microscopically, mev/mev mice revealed scarce and large prostatic ducts, acini severely atrophic with empty lumen and scarce loose epithelial component forming tufts and infoldings, and hyperplastic changes in fibromuscular stroma. CONCLUSIONS The prostate of mev/mev mice exhibits signs of aberrant differentiation and the resulting phenotype may be related to the loss of function of SHP-1. Prostatic anomalies in these mice affect, together with defects in sperm maduration, for their sterility. These data suggest SHP-1 plays an important role in prostate epithelial morphogenesis.
Collapse
|
32
|
Kumar S, Xu J, Kumar RS, Lakshmikanthan S, Kapur R, Kofron M, Chrzanowska-Wodnicka M, Filippi MD. The small GTPase Rap1b negatively regulates neutrophil chemotaxis and transcellular diapedesis by inhibiting Akt activation. ACTA ACUST UNITED AC 2014; 211:1741-58. [PMID: 25092872 PMCID: PMC4144729 DOI: 10.1084/jem.20131706] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Mice lacking the small GTPase Rap1b exhibit enhanced neutrophil recruitment to inflamed lungs and susceptibility to endotoxin shock via enhance PI3K-Akt activation. Neutrophils are the first line of cellular defense in response to infections and inflammatory injuries. However, neutrophil activation and accumulation into tissues trigger tissue damage due to release of a plethora of toxic oxidants and proteases, a cause of acute lung injury (ALI). Despite its clinical importance, the molecular regulation of neutrophil migration is poorly understood. The small GTPase Rap1b is generally viewed as a positive regulator of immune cell functions by controlling bidirectional integrin signaling. However, we found that Rap1b-deficient mice exhibited enhanced neutrophil recruitment to inflamed lungs and enhanced susceptibility to endotoxin shock. Unexpectedly, Rap1b deficiency promoted the transcellular route of diapedesis through endothelial cell. Increased transcellular migration of Rap1b-deficient neutrophils in vitro was selectively mediated by enhanced PI3K-Akt activation and invadopodia-like protrusions. Akt inhibition in vivo suppressed excessive Rap1b-deficient neutrophil migration and associated endotoxin shock. The inhibitory action of Rap1b on PI3K signaling may be mediated by activation of phosphatase SHP-1. Thus, this study reveals an unexpected role for Rap1b as a key suppressor of neutrophil migration and lung inflammation.
Collapse
Affiliation(s)
- Sachin Kumar
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229 University of Cincinnati College of Medicine, Cincinnati OH 45229
| | - Juying Xu
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229 University of Cincinnati College of Medicine, Cincinnati OH 45229
| | - Rupali Sani Kumar
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229 University of Cincinnati College of Medicine, Cincinnati OH 45229
| | | | - Reuben Kapur
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Cancer Research Institute, Indianapolis, IN 46202
| | - Matthew Kofron
- Division of Developmental Biology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229
| | | | - Marie-Dominique Filippi
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229 University of Cincinnati College of Medicine, Cincinnati OH 45229
| |
Collapse
|
33
|
Marín-Juez R, Jong-Raadsen S, Yang S, Spaink HP. Hyperinsulinemia induces insulin resistance and immune suppression via Ptpn6/Shp1 in zebrafish. J Endocrinol 2014; 222:229-41. [PMID: 24904114 DOI: 10.1530/joe-14-0178] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Type 2 diabetes, obesity, and metabolic syndrome are pathologies where insulin resistance plays a central role, and that affect a large population worldwide. These pathologies are usually associated with a dysregulation of insulin secretion leading to a chronic exposure of the tissues to high insulin levels (i.e. hyperinsulinemia), which diminishes the concentration of key downstream elements, causing insulin resistance. The complexity of the study of insulin resistance arises from the heterogeneity of the metabolic states where it is observed. To contribute to the understanding of the mechanisms triggering insulin resistance, we have developed a zebrafish model to study insulin metabolism and its associated disorders. Zebrafish larvae appeared to be sensitive to human recombinant insulin, becoming insulin-resistant when exposed to a high dose of the hormone. Moreover RNA-seq-based transcriptomic profiling of these larvae revealed a strong downregulation of a number of immune-relevant genes as a consequence of the exposure to hyperinsulinemia. Interestingly, as an exception, the negative immune modulator protein tyrosine phosphatase nonreceptor type 6 (ptpn6) appeared to be upregulated in insulin-resistant larvae. Knockdown of ptpn6 was found to counteract the observed downregulation of the immune system and insulin signaling pathway caused by hyperinsulinemia. These results indicate that ptpn6 is a mediator of the metabolic switch between insulin-sensitive and insulin-resistant states. Our zebrafish model for hyperinsulinemia has therefore demonstrated its suitability for discovery of novel regulators of insulin resistance. In addition, our data will be very useful in further studies of the function of immunological determinants in a non-obese model system.
Collapse
Affiliation(s)
- Rubén Marín-Juez
- ZF-Screens BVJ.H. Oortweg 19, 2333 CH Leiden, The NetherlandsInstitute of BiologyLeiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Susanne Jong-Raadsen
- ZF-Screens BVJ.H. Oortweg 19, 2333 CH Leiden, The NetherlandsInstitute of BiologyLeiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Shuxin Yang
- ZF-Screens BVJ.H. Oortweg 19, 2333 CH Leiden, The NetherlandsInstitute of BiologyLeiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Herman P Spaink
- ZF-Screens BVJ.H. Oortweg 19, 2333 CH Leiden, The NetherlandsInstitute of BiologyLeiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| |
Collapse
|
34
|
Becerra-Díaz M, Terrazas LI. Taenia crassiceps infection and its excreted/secreted products inhibit STAT1 activation in response to IFN-γ. Int J Parasitol 2014; 44:613-23. [PMID: 24837622 DOI: 10.1016/j.ijpara.2014.03.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 03/21/2014] [Accepted: 03/25/2014] [Indexed: 01/08/2023]
Abstract
It is well understood that helminth infections modulate the immune responses of their hosts but the mechanisms involved in this modulation are not fully known. Macrophages and dendritic cells appear to be consistently affected during this type of infection and are common target cells for helminth-derived molecules. In this report, we show that macrophages obtained from chronically Taenia crassiceps-infected mice displayed an impaired response to recombinant murine IFN-γ, but not to recombinant murine IL-4, as measured based on the phosphorylation of STAT1 and STAT6, respectively. These macrophages expressed high levels of SOCS3. However, the inhibition of phosphatase activity by orthovanadate restored the IFN-γ response of these macrophages by increasing STAT1 phosphorylation without affecting SOCS3 expression. Therefore, we aimed to identify the phosphatases associated with IFN-γ signaling inhibition and found that macrophages from T. crassiceps-infected mice displayed enhanced SHP-1 expression. Interestingly, the exposure of naïve macrophages to T. crassiceps excreted/secreted products similarly interfered with IFN-γ-induced STAT1 phosphorylation. Moreover, macrophages exposed to T. crassiceps excreted/secreted products expressed high levels of SOCS3 as well as SHP-1. Strikingly, human peripheral blood mononuclear cells that were exposed to T. crassiceps excreted/secreted products in vitro also displayed impaired STAT1 phosphorylation in response to IFN-γ; again, phosphatase inhibition abrogated the T. crassiceps excreted/secreted product-altered IFN-γ signaling. These data demonstrate a new mechanism by which helminth infection and the products derived during this infection target intracellular pathways to block the response to inflammatory cytokines such as IFN-γ in both murine and human cells.
Collapse
Affiliation(s)
- Mireya Becerra-Díaz
- Unidad de Biomedicina, Facultad de Estudios Superiores (FES)-Iztacala, Universidad Nacional Autónoma de México (UNAM), Av. De los Barrios 1, Los Reyes Iztacala, Tlalnepantla, Edo. de Mexico 54090, Mexico
| | - Luis I Terrazas
- Unidad de Biomedicina, Facultad de Estudios Superiores (FES)-Iztacala, Universidad Nacional Autónoma de México (UNAM), Av. De los Barrios 1, Los Reyes Iztacala, Tlalnepantla, Edo. de Mexico 54090, Mexico.
| |
Collapse
|
35
|
Evren S, Wan S, Ma XZ, Fahim S, Mody N, Sakac D, Jin T, Branch DR. Characterization of SHP-1 protein tyrosine phosphatase transcripts, protein isoforms and phosphatase activity in epithelial cancer cells. Genomics 2013; 102:491-9. [DOI: 10.1016/j.ygeno.2013.10.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 09/26/2013] [Accepted: 10/01/2013] [Indexed: 10/26/2022]
|
36
|
Witzig TE, Hu G, Offer SM, Wellik LE, Han JJ, Stenson MJ, Dogan A, Diasio RB, Gupta M. Epigenetic mechanisms of protein tyrosine phosphatase 6 suppression in diffuse large B-cell lymphoma: implications for epigenetic therapy. Leukemia 2013; 28:147-54. [PMID: 23979523 DOI: 10.1038/leu.2013.251] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 08/07/2013] [Accepted: 08/16/2013] [Indexed: 11/09/2022]
Abstract
Protein tyrosine phosphatases such as PTPN6 can be downregulated in various neoplasms. PTPN6 expression by immunohistochemistry in 40 diffuse large B-cell lymphoma (DLBCL) tumors was lost or suppressed in 53% (21/40). To elucidate the molecular mechanisms of PTPN6 suppression, we performed a comprehensive epigenetic analysis of PTPN6 promoter 2 (P2). None of the DLBCL primary tumors (0/37) had PTPN6 hypermethylation on the CpG1 island using methylation-specific PCR, pyrosequencing, and high-resolution melting assays. However, hypermethylation in 57% (21/37) of cases was found in a novel CpG island (CpG2) in P2. PTPN6 gene suppression was reversed by 5-aza-deoxycytidine (5-Aza), a DNA methyltransferase inhibitor, and the histone deacetylase inhibitor (HDACi) LBH589. LBH589 and 5-Aza in combination inhibited DLBCL survival and PTPN6 hypermethylation at CpG2. The role of histone modifications was investigated with a chromatin-immunoprecipitation assay demonstrating that PTPN6 P2 is associated with silencing histone marks H3K27me3 and H3K9me3 in DLBCL cells but not normal B cells. 3-Deazaneplanocin A, a histone methyltransferase inhibitor, decreased the H3K27me3 mark, whereas HDACi LBH589 increased the H3K9Ac mark within P2 resulting in re-expression of PTPN6. These studies have uncovered novel epigenetic mechanisms of PTPN6 suppression and suggest that PTPN6 may be a potential target of epigenetic therapy in DLBCL.
Collapse
Affiliation(s)
- T E Witzig
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - G Hu
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - S M Offer
- Department of Molecular and Experimental Therapeutics, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - L E Wellik
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - J J Han
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - M J Stenson
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - A Dogan
- Department of Pathology, Memorial Sloan-Kettering Cancer, New York, NY, USA
| | - R B Diasio
- Department of Molecular and Experimental Therapeutics, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - M Gupta
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, College of Medicine, Rochester, MN, USA
| |
Collapse
|
37
|
Okenwa C, Kumar A, Rego D, Konarski Y, Nilchi L, Wright K, Kozlowski M. SHP-1-Pyk2-Src protein complex and p38 MAPK pathways independently regulate IL-10 production in lipopolysaccharide-stimulated macrophages. THE JOURNAL OF IMMUNOLOGY 2013; 191:2589-603. [PMID: 23904162 DOI: 10.4049/jimmunol.1300466] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The role of tyrosine phosphatase Src homology region 2 domain-containing phosphatase (SHP)-1 in LPS-activated cytokine production and inflammation was investigated by determining TNF-α and IL-10 production in splenic macrophages employing SHP-1-null (me/me) mouse model. LPS-stimulated me/me splenic macrophages secreted significantly less IL-10 with concomitantly elevated levels of TNF-α compared with wild-type (WT) macrophages irrespective of LPS dose and duration of stimulation. IL-10 significantly inhibited LPS-induced TNF-α production in both me/me and WT macrophages. The critical requirement for SHP-1 in regulating LPS-induced IL-10 and TNF-α production was confirmed by interfering with SHP-1 expression in WT macrophages and by reconstituting me/me macrophages with the SHP-1 gene. To delineate the role of SHP-1 in positive regulation of LPS-induced IL-10 production, signaling proteins representing SHP-1 targets were examined. The results reveal that tyrosine kinases Src and proline-rich tyrosine kinase 2 (Pyk2) regulate SHP-1-dependent LPS-induced IL-10 production and infer that optimal LPS-induced IL-10 production requires an assembly of a protein complex consisting of SHP-1-Pyk2-Src proteins. Moreover, LPS-induced IL-10 production also requires activation of the p38 MAPK independent of SHP-1 function. Overall, to our knowledge our results show for the first time that SHP-1 acts as a positive regulator of LPS-induced IL-10 production in splenic macrophages through two distinct and independent SHP-1-Pyk2-Src and p38 MAPK pathways.
Collapse
Affiliation(s)
- Chinonso Okenwa
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | | | | | | | | | | | | |
Collapse
|
38
|
Kawakami T, Xiao W, Yasudo H, Kawakami Y. Regulation of proliferation, survival, differentiation, and activation by the Signaling Platform for SHP-1 phosphatase. Adv Biol Regul 2013; 52:7-15. [PMID: 21982978 DOI: 10.1016/j.advenzreg.2011.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Accepted: 09/05/2011] [Indexed: 12/20/2022]
Affiliation(s)
- Toshiaki Kawakami
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA.
| | | | | | | |
Collapse
|
39
|
Jung JH, Jeong SJ, Kim JH, Jung SK, Jung DB, Lee D, Sohn EJ, Yun M, Lee HJ, Lee HJ, Kim SH. Inactivation of HDAC3 and STAT3 is Critically Involved in 1-Stearoyl-sn-Glycero-3-Phosphocholine-Induced Apoptosis in Chronic Myelogenous Leukemia K562 Cells. Cell Biochem Biophys 2013; 67:1379-89. [DOI: 10.1007/s12013-013-9670-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
40
|
Abram CL, Roberge GL, Pao LI, Neel BG, Lowell CA. Distinct roles for neutrophils and dendritic cells in inflammation and autoimmunity in motheaten mice. Immunity 2013; 38:489-501. [PMID: 23521885 DOI: 10.1016/j.immuni.2013.02.018] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Accepted: 11/26/2012] [Indexed: 01/18/2023]
Abstract
The motheaten mouse has long served as a paradigm for complex autoimmune and inflammatory disease. Null mutations in Ptpn6, which encodes the nonreceptor protein-tyrosine phosphatase Shp1, cause the motheaten phenotype. However, Shp1 regulates multiple signaling pathways in different hematopoietic cell types, so the cellular and molecular mechanism of autoimmunity and inflammation in the motheaten mouse has remained unclear. By using floxed Ptpn6 mice, we dissected the contribution of innate immune cells to the motheaten phenotype. Ptpn6 deletion in neutrophils resulted in cutaneous inflammation, but not autoimmunity, providing an animal model of human neutrophilic dermatoses. By contrast, dendritic cell deletion caused severe autoimmunity, without inflammation. Genetic and biochemical analysis showed that inflammation was caused by enhanced neutrophil integrin signaling through Src-family and Syk kinases, whereas autoimmunity resulted from exaggerated MyD88-dependent signaling in dendritic cells. Our data demonstrate that disruption of distinct Shp1-regulated pathways in different cell types combine to cause motheaten disease.
Collapse
Affiliation(s)
- Clare L Abram
- Department of Laboratory Medicine and the Program in Immunology, University of California, San Francisco 94143, USA
| | | | | | | | | |
Collapse
|
41
|
Kanwal Z, Zakrzewska A, den Hertog J, Spaink HP, Schaaf MJM, Meijer AH. Deficiency in hematopoietic phosphatase ptpn6/Shp1 hyperactivates the innate immune system and impairs control of bacterial infections in zebrafish embryos. THE JOURNAL OF IMMUNOLOGY 2013; 190:1631-45. [PMID: 23335748 DOI: 10.4049/jimmunol.1200551] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Deficiency in Src homology region 2 domain-containing phosphatase 1/protein tyrosine phosphatase nonreceptor type 6 (SHP1/PTPN6) is linked with chronic inflammatory diseases and hematological malignancies in humans. In this study, we exploited the embryonic and larval stages of zebrafish (Danio rerio) as an animal model to study ptpn6 function in the sole context of innate immunity. We show that ptpn6 knockdown induces a spontaneous inflammation-associated phenotype at the late larval stage. Surprisingly, glucocorticoid treatment did not suppress inflammation under ptpn6 knockdown conditions but further enhanced leukocyte infiltration and proinflammatory gene expression. Experiments in a germ-free environment showed that the late larval phenotype was microbe independent. When ptpn6 knockdown embryos were challenged with Salmonella typhimurium or Mycobacterium marinum at earlier stages of development, the innate immune system was hyperactivated to a contraproductive level that impaired the control of these pathogenic bacteria. Transcriptome analysis demonstrated that Kyoto Encyclopedia of Genes and Genomes pathways related to pathogen recognition and cytokine signaling were significantly enriched under these conditions, suggesting that ptpn6 functions as a negative regulator that imposes a tight control over the level of innate immune response activation during infection. In contrast to the hyperinduction of proinflammatory cytokine genes under ptpn6 knockdown conditions, anti-inflammatory il10 expression was not hyperinduced. These results support that ptpn6 has a crucial regulatory function in preventing host-detrimental effects of inflammation and is essential for a successful defense mechanism against invading microbes.
Collapse
Affiliation(s)
- Zakia Kanwal
- Institute of Biology, Leiden University, 2333 CC Leiden, The Netherlands
| | | | | | | | | | | |
Collapse
|
42
|
Van Dyken SJ, Locksley RM. Interleukin-4- and interleukin-13-mediated alternatively activated macrophages: roles in homeostasis and disease. Annu Rev Immunol 2013; 31:317-43. [PMID: 23298208 DOI: 10.1146/annurev-immunol-032712-095906] [Citation(s) in RCA: 535] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The macrophage, a versatile cell type prominently involved in host defense and immunity, assumes a distinct state of alternative activation in the context of polarized type 2 immune responses such as allergic inflammation and helminth infection. This alternatively activated phenotype is induced by the canonical type 2 cytokines interleukin (IL)-4 and IL-13, which mediate expression of several characteristic markers along with a dramatic shift in macrophage metabolic pathways that influence surrounding cells and tissues. We discuss recent advances in the understanding of IL-4- and IL-13-mediated alternatively activated macrophages and type 2 immune responses; such advances have led to an expanded appreciation for functions of these cells beyond immunity, including maintenance of physiologic homeostasis and tissue repair.
Collapse
Affiliation(s)
- Steven J Van Dyken
- Departments of Medicine and Microbiology & Immunology, Howard Hughes Medical Institute, University of California, San Francisco, California 94143, USA
| | | |
Collapse
|
43
|
Stanford SM, Rapini N, Bottini N. Regulation of TCR signalling by tyrosine phosphatases: from immune homeostasis to autoimmunity. Immunology 2012; 137:1-19. [PMID: 22862552 DOI: 10.1111/j.1365-2567.2012.03591.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
More than half of the known protein tyrosine phosphatases (PTPs) in the human genome are expressed in T cells, and significant progress has been made in elucidating the biology of these enzymes in T-cell development and function. Here we provide a systematic review of the current understanding of the roles of PTPs in T-cell activation, providing insight into their mechanisms of action and regulation in T-cell receptor signalling, the phenotypes of their genetically modified mice, and their possible involvement in T-cell-mediated autoimmune disease. Our projection is that the interest in PTPs as mediators of T-cell homeostasis will continue to rise with further functional analysis of these proteins, and PTPs will be increasingly considered as targets of immunomodulatory therapies.
Collapse
Affiliation(s)
- Stephanie M Stanford
- Division of Cellular Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
44
|
Liu SH, Wang KB, Lan KH, Lee WJ, Pan HC, Wu SM, Peng YC, Chen YC, Shen CC, Cheng HC, Liao KK, Sheu ML. Calpain/SHP-1 interaction by honokiol dampening peritoneal dissemination of gastric cancer in nu/nu mice. PLoS One 2012; 7:e43711. [PMID: 22937084 PMCID: PMC3427156 DOI: 10.1371/journal.pone.0043711] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 07/24/2012] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Honokiol, a small-molecular weight natural product, has previously been reported to activate apoptosis and inhibit gastric tumorigenesis. Whether honokiol inhibits the angiogenesis and metastasis of gastric cancer cells remains unknown. METHODOLOGY/PRINCIPAL FINDINGS We tested the effects of honokiol on angiogenic activity and peritoneal dissemination using in vivo, ex vivo and in vitro assay systems. The signaling responses in human gastric cancer cells, human umbilical vascular endothelial cells (HUVECs), and isolated tumors were detected and analyzed. In a xenograft gastric tumor mouse model, honokiol significantly inhibited the peritoneal dissemination detected by PET/CT technique. Honokiol also effectively attenuated the angiogenesis detected by chick chorioallantoic membrane assay, mouse matrigel plug assay, rat aortic ring endothelial cell sprouting assay, and endothelial cell tube formation assay. Furthermore, honokiol effectively enhanced signal transducer and activator of transcription (STAT-3) dephosphorylation and inhibited STAT-3 DNA binding activity in human gastric cancer cells and HUVECs, which was correlated with the up-regulation of the activity and protein expression of Src homology 2 (SH2)-containing tyrosine phosphatase-1 (SHP-1). Calpain-II inhibitor and siRNA transfection significantly reversed the honokiol-induced SHP-1 activity. The decreased STAT-3 phosphorylation and increased SHP-1 expression were also shown in isolated peritoneal metastatic tumors. Honokiol was also capable of inhibiting VEGF generation, which could be reversed by SHP-1 siRNA transfection. CONCLUSIONS/SIGNIFICANCE Honokiol increases expression and activity of SPH-1 that further deactivates STAT3 pathway. These findings also suggest that honokiol is a novel and potent inhibitor of angiogenesis and peritoneal dissemination of gastric cancer cells, providing support for the application potential of honokiol in gastric cancer therapy.
Collapse
Affiliation(s)
- Shing Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Keh Bin Wang
- Department of Nuclear Medicine, Kuang Tien General Hospital, Taichung, Taiwan
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Keng Hsin Lan
- Division of Gastroenterology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Wen Jane Lee
- Department of Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Hung Chuan Pan
- Department of Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Sheng Mao Wu
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Yen Chun Peng
- Division of Gastroenterology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yi Ching Chen
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Chin Chang Shen
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Hsu Chen Cheng
- Department of life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Ko Kaung Liao
- Department of Anatomy, Chung Shan Medical University, Taichung, Taiwan
| | - Meei Ling Sheu
- Department of Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
- * E-mail:
| |
Collapse
|
45
|
Draber P, Stepanek O, Hrdinka M, Drobek A, Chmatal L, Mala L, Ormsby T, Angelisova P, Horejsi V, Brdicka T. LST1/A is a myeloid leukocyte-specific transmembrane adaptor protein recruiting protein tyrosine phosphatases SHP-1 and SHP-2 to the plasma membrane. J Biol Chem 2012; 287:22812-21. [PMID: 22589543 DOI: 10.1074/jbc.m112.339143] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Transmembrane adaptor proteins are membrane-anchored proteins consisting of a short extracellular part, a transmembrane domain, and a cytoplasmic part with various protein-protein interaction motifs but lacking any enzymatic activity. They participate in the regulation of various signaling pathways by recruiting other proteins to the proximity of cellular membranes where the signaling is often initiated and propagated. In this work, we show that LST1/A, an incompletely characterized protein encoded by MHCIII locus, is a palmitoylated transmembrane adaptor protein. It is expressed specifically in leukocytes of the myeloid lineage, where it localizes to the tetraspanin-enriched microdomains. In addition, it binds SHP-1 and SHP-2 phosphatases in a phosphotyrosine-dependent manner, facilitating their recruitment to the plasma membrane. These data suggest a role for LST1/A in negative regulation of signal propagation.
Collapse
Affiliation(s)
- Peter Draber
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, 142 20 Prague, Czech Republic
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Capello D, Gloghini A, Baldanzi G, Martini M, Deambrogi C, Lucioni M, Piranda D, Famà R, Graziani A, Spina M, Tirelli U, Paulli M, Larocca LM, Gaidano G, Carbone A, Sinigaglia F. Alterations of negative regulators of cytokine signalling in immunodeficiency-related non-Hodgkin lymphoma. Hematol Oncol 2012; 31:22-8. [PMID: 22488585 DOI: 10.1002/hon.2010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 02/07/2012] [Accepted: 02/20/2012] [Indexed: 12/19/2022]
Abstract
We investigated immunodeficiency-related non-Hodgkin lymphoma for the presence of molecular alterations affecting negative regulators of the Janus family protein tyrosine kinase/signal transducer and activator of transcription pathway. Protein tyrosine phosphatase, non-receptor type 6/Src homology 2-containing tyrosine phosphatase-1 epigenetic silencing was recurrent in primary effusion lymphoma (100%), and diffuse large B-cell lymphoma (63%), with a higher prevalence in the non-germinal centre subtype, and was associated with the activation of the Janus family protein tyrosine kinase/signal transducer and activator of transcription 3 pathway. Suppressor of cytokine signalling (SOCS)1 and SOCS3 epigenetic silencing were occasionally detected, whereas SOCS1 was frequently mutated in diffuse large B-cell lymphoma and polymorphic post-transplant lymphoproliferative disorders, possibly as a cause of aberrant somatic hypermutation. However, the mutation profile of the coding region of the gene was different from that expected from the aberrant somatic hypermutation process, suggesting that, at least in some cases, SOCS1 mutations may have been selected for their functional activity.
Collapse
Affiliation(s)
- Daniela Capello
- Division of Hematology, 'Amedeo Avogadro' University of Eastern Piedmont, Novara, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
TGF-β1 re-programs TLR4 signaling in L. donovani infection: enhancement of SHP-1 and ubiquitin-editing enzyme A20. Immunol Cell Biol 2011; 90:640-54. [PMID: 21968712 DOI: 10.1038/icb.2011.80] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Visceral leishmaniasis (VL), caused by Leishmania donovani, is a major health concern in India. It represents T-helper type 2 (Th2) bias of cytokines in active state and Th1 bias at cure. However, the role of the parasite in regulating Toll-like receptor (TLR)-mediated macrophage activation in VL patients remains elusive. In this report, we demonstrated that later stages of L. donovani infection rendered tolerance to macrophages, leading to incapability for the production of inflammatory cytokines like tumor necrosis factor (TNF)-α and interleukin (IL)-1β in response to TLR stimulation. Overexpression of transforming growth factor (TGF)-β(1), but not IL-10, resulted in suppressed lipopolysaccharide (LPS)-induced production of TNF-α and downregulation of TLR4 expression in L. donovani-infected macrophages. Recombinant human (rh)TGF-β(1) markedly enhanced tyrosine phosphatase (Src homology region 2 domain-containing phosphatase-1) activity, but inhibited IL-1 receptor-activated kinase (IRAK)-1 activation. Addition of neutralizing TGF-β(1) antibody reversed these effects, and thus suggesting the pivotal role of TGF-β(1) in promoting refractoriness for LPS in macrophages. Surprisingly, the use of a tyrosine phosphatase inhibitor (sodium orthovanadate, Na(3)VO(4)) promoted IRAK-1 activation, confirming the negative inhibitory role of tyrosine phosphatase in macrophage activation. Furthermore, rhTGF-β(1) induced tolerance in infected macrophages by reducing inhibitory protein (IκBα) degradation in a time-dependent manner. In addition, short interfering RNA studies proved that overexpression of A20 ubiquitin-editing protein complex induced inhibitory activity of TGF-β(1) on LPS-mediated nuclear factor-κB activation. Thus, these findings suggest that TGF-β(1) promotes overexpression of A20 through tyrosine phosphatase activity that ensures transient activation of inflammatory signaling pathways in macrophages in active L. donovani infection.
Collapse
|
48
|
Amin S, Kumar A, Nilchi L, Wright K, Kozlowski M. Breast cancer cells proliferation is regulated by tyrosine phosphatase SHP1 through c-jun N-terminal kinase and cooperative induction of RFX-1 and AP-4 transcription factors. Mol Cancer Res 2011; 9:1112-25. [PMID: 21719561 DOI: 10.1158/1541-7786.mcr-11-0097] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In this study, we show that proliferation of breast cancer cells is suppressed by IGF-1-activated JNK MAPK pathway. The molecular mechanism by which c-jun-NH,-kinase (JNK) activation induces antiproliferative signals in IGF-1-stimulated breast cancer cells remains unknown. Tyrosine phosphatase SHP1 is known to negatively regulate signal transduction pathways activated by cell surface receptors including IGF-1. Moreover, SHP1 transcript and protein levels are increased in epithelial tumors. Therefore, we hypothesized that IGF-activated JNK induces expression of SHP1 in breast cancer cells. To further clarify the role of SHP1 in tumor growth, we correlated the proliferation rates of breast adenocarcinoma cells with SHP1 expression and JNK activation. We show that proliferation of serum- or IGF-1-stimulated breast adenocarcinoma cells is negatively regulated by SHP1 and show for the first time that IGF-1-activated JNK induces SHP1 expression in MCF-7 cells used as experimental model. In an attempt to understand the mechanism by which serum- or IGF-1-activated JNK induces SHP1 expression resulting in suppression of cell proliferation, we reveal for the first time that in serum- or IGF-1-stimulated breast cancer MCF-7 cells, JNK induces SHP1 expression through the binding of AP-4 and RFX-1 transcription factors to the epithelial tissue-specific SHP1 promoter. Overall, we show for the first time that IGF-1-stimulated proliferation of breast adenocarcinoma cells is negatively regulated by SHP1 through activation of JNK.
Collapse
Affiliation(s)
- Shahreen Amin
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | | | | | | | | |
Collapse
|
49
|
Rego D, Kumar A, Nilchi L, Wright K, Huang S, Kozlowski M. IL-6 Production Is Positively Regulated by Two DistinctSrcHomology Domain 2-Containing Tyrosine Phosphatase-1 (SHP-1)–Dependent CCAAT/Enhancer-Binding Protein β and NF-κB Pathways and an SHP-1–Independent NF-κB Pathway in Lipopolysaccharide-Stimulated Bone Marrow-Derived Macrophages. THE JOURNAL OF IMMUNOLOGY 2011; 186:5443-56. [DOI: 10.4049/jimmunol.1003551] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
50
|
Ramachandran IR, Song W, Lapteva N, Seethammagari M, Slawin KM, Spencer DM, Levitt JM. The phosphatase SRC homology region 2 domain-containing phosphatase-1 is an intrinsic central regulator of dendritic cell function. THE JOURNAL OF IMMUNOLOGY 2011; 186:3934-45. [PMID: 21357539 DOI: 10.4049/jimmunol.1001675] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dendritic cells (DCs) initiate proinflammatory or regulatory T cell responses, depending on their activation state. Despite extensive knowledge of DC-activating signals, the understanding of DC inhibitory signals is relatively limited. We show that Src homology region 2 domain-containing phosphatase-1 (SHP-1) is an important inhibitor of DC signaling, targeting multiple activation pathways. Downstream of TLR4, SHP-1 showed increased interaction with several proteins including IL-1R-associated kinase-4, and modulated LPS signaling by inhibiting NF-κB, AP-1, ERK, and JNK activity, while enhancing p38 activity. In addition, SHP-1 inhibited prosurvival signaling through AKT activation. Furthermore, SHP-1 inhibited CCR7 protein expression. Inhibiting SHP-1 in DCs enhanced proinflammatory cytokines, IL-6, IL-12, and IL-1β production, promoted survival, and increased DC migration to draining lymph nodes. Administration of SHP-1-inhibited DCs in vivo induced expansion of Ag-specific cytotoxic T cells and inhibited Foxp3(+) regulatory T cell induction, resulting in an enhanced immune response against pre-established mouse melanoma and prostate tumors. Taken together, these data demonstrate that SHP-1 is an intrinsic global regulator of DC function, controlling many facets of T cell-mediated immune responses.
Collapse
Affiliation(s)
- Indu R Ramachandran
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | | | |
Collapse
|