1
|
Hopke A, Viens AL, Alexander NJ, Mun SJ, Mansour MK, Irimia D. Spleen tyrosine kinase inhibitors disrupt human neutrophil swarming and antifungal functions. Microbiol Spectr 2025; 13:e0254921. [PMID: 39601545 PMCID: PMC11705959 DOI: 10.1128/spectrum.02549-21] [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: 12/09/2021] [Accepted: 10/26/2024] [Indexed: 11/29/2024] Open
Abstract
Neutrophils communicate with one another and amplify their destructive power through swarming, a collective process that synchronizes the activities of multiple neutrophils against one target. The sequence of activities contributing to swarming against clusters of fungi has been recently uncovered. However, the molecular signals controlling the neutrophils' activities during the swarming process are just emerging. Here, we report that spleen tyrosine kinase (SYK) inhibitors severely impair neutrophil swarming responses, resulting in the complete loss of fungal restriction. These findings are enabled by a microscale platform to probe the biology of human neutrophils swarming against uniformly sized clusters of growing Candida albicans, a representative opportunistic fungal pathogen. We take advantage of the ability to monitor large arrays of swarms and quantify the effect of multiple chemical inhibitors on different phases of human neutrophil swarming. We show that inhibitors that interfere with PI3Ky signaling disrupt the regulation of the initiation of swarming, while the activation of JNK signaling is essential for the activation of biochemical antifungal functions. Furthermore, we reveal that granulocyte colony-stimulating factors (GCSF and GM-CSF) can partially rescue the antifungal functions of neutrophils exposed to SYK inhibitors. These findings advance our understanding of neutrophil swarming biology in humans and lay the foundation for novel therapeutics that may restore neutrophil function during immunosuppression. IMPORTANCE Neutrophils can amplify their destructive power through swarming, a crucial process against large targets that individual neutrophils cannot destroy. However, the molecular mechanisms controlling this process are just emerging. Here, we leveraged microscale tools to probe the biology of swarming against fungi. We used multiple chemical inhibitors and mapped SYK, PI3Ky, and JNK signaling roles during human neutrophil swarming against fungal clusters of Candida albicans. We also found that treating human neutrophils with GCSF and GM-CSF rescues some neutrophil antifungal function during SYK inhibition. These findings advance our understanding of swarming biology in humans while laying the foundation for developing therapeutics that enhance neutrophil function during immunosuppression.
Collapse
Affiliation(s)
- Alex Hopke
- BioMEMS Resource Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Shriners Hospital for Children, Boston, Massachusetts, USA
| | - Adam L. Viens
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Natalie J. Alexander
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Seok Joon Mun
- BioMEMS Resource Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Michael K. Mansour
- Harvard Medical School, Boston, Massachusetts, USA
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Daniel Irimia
- BioMEMS Resource Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Shriners Hospital for Children, Boston, Massachusetts, USA
| |
Collapse
|
2
|
Jia W, Luo S, Guo H, Kong D. Development of PI3Kα inhibitors for tumor therapy. J Biomol Struct Dyn 2023; 41:8587-8604. [PMID: 36221910 DOI: 10.1080/07391102.2022.2132293] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 09/28/2022] [Indexed: 10/17/2022]
Abstract
The PI3K/AKT/mTOR signaling pathway is well known to be involved in cell growth, proliferation, metabolism and other cellular physiological processes. Abnormal activation of this pathway is closely related to tumorigenesis and metastasis. As the starting node of the pathway, PI3K is known to contain 4 isoforms, including PI3Kα, a heterodimer composed of the catalytic subunit p110α and the regulatory subunit p85. PIK3CA, which encodes p110α, is frequently mutated in cancer, especially breast cancer. Abnormal activation of PI3Kα promotes cancer cell proliferation, migration, invasion, and angiogenesis; therefore, PI3Kα has become a key target for the development of anticancer drugs. The hinge region and the region of the mutation site in the PI3Kα protein are important for designing PI3Kα-specific inhibitors. As the group shared by the most PI3Kα-specific inhibitors reported thus far, carboxamide can produce hydrogen bonds with Gln859 and Ser854. Gln859 is specific to the p110α protein in producing hydrogen bond interactions with PI3Kα-specific inhibitors and this is a key point for designing PI3Kα inhibitors. To date, alpelisib is the only PI3Kα inhibitor approved for the treatment of breast cancer. Several other PI3Kα inhibitors are under evaluation in clinical trials. In this review, we briefly describe PI3Kα and its role in tumorigenesis, summarize the clinical trial results of some PI3Kα inhibitors as well as the synthetic routes of alpelisib, and finally give our proposal for the development of novel PI3Kα inhibitors for tumor therapy. HighlightsWe summarize the progress of PI3Kα and PI3Kα inhibitors in cancer from the second half of the 20th century to the present.We describe the clinical trial results of PI3Kα inhibitors as well as the synthetic routes of the only approved PI3Kα inhibitor alpelisib.Crystal structure of alpelisib bound to the PI3Kα receptor binding domain.This review gives proposal for the development of novel PI3Kα inhibitors and will serve as a complementary summary to other reviews in the research field of PI3K inhibitors.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Wenqing Jia
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Shuyu Luo
- School of Stomatology, Hospital of Stomatology, Tianjin Medical University, Tianjin, China
| | - Han Guo
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Dexin Kong
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| |
Collapse
|
3
|
Geng R, Huang X, Li L, Guo X, Wang Q, Zheng Y, Guo X. Gene expression analysis in endometriosis: Immunopathology insights, transcription factors and therapeutic targets. Front Immunol 2022; 13:1037504. [PMID: 36532015 PMCID: PMC9748153 DOI: 10.3389/fimmu.2022.1037504] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/15/2022] [Indexed: 12/05/2022] Open
Abstract
Background Endometriosis is recognized as an estrogen-dependent inflammation disorder, estimated to affect 8%-15% of women of childbearing age. Currently, the etiology and pathogenesis of endometriosis are not completely clear. Underlying mechanism for endometriosis is still under debate and needs further exploration. The involvement of transcription factors and immune mediations may be involved in the pathophysiological process of endometriosis, but the specific mechanism remains to be explored. This study aims to investigate the underlying molecular mechanisms in endometriosis. Methods The gene expression profile of endometriosis was obtained from the gene expression omnibus (GEO) database. Gene set variation analysis (GSVA) and gene set enrichment analysis (GSEA) were applied to the endometriosis GSE7305 datasets. Cibersort and MCP-counter were used to explore the immune response gene sets, immune response pathway, and immune environment. Differentially expressed genes (DEGs) were identified and screened. Common biological pathways were being investigated using the kyoto encyclopedia of genes and genomes (KEGG) pathway enrichment analysis. Transcription factors were from The Human Transcription Factors. The least absolute shrinkage and selection operator (Lasso) model identified four differential expressions of transcription factors (AEBP1, HOXB6, KLF2, and RORB). Their diagnostic value was calculated by receiver operating characteristic (ROC) curve analysis and validated in the validation cohort (GSE11691, GSE23339). By constructing the interaction network of crucial transcription factors, weighted gene coexpression network analysis (WGCNA) was used to search for key module genes. Metascape was used for enrichment analysis of essential module genes and obtained HOXB6, KLF2. The HOXB6 and KLF2 were further verified as the only two intersection genes according to Support Vector Machine Recursive Feature Elimination (SVM-RFE) and random forest models. We constructed ceRNA (lncRNA-miRNA-mRNA) networks with four potential transcription factors. Finally, we performed molecular docking for goserelin and dienogest with four transcription factors (AEBP1, HOXB6, KLF2, and RORB) to screen potential drug targets. Results Immune and metabolic pathways were enriched in GSVA and GSEA. In single sample gene set enrichment analysis (ssGSEA), most immune infiltrating cells, immune response gene sets, and immune response pathways are differentially expressed between endometriosis and non-endometriosis. Twenty-seven transcription factors were screened from differentially expressed genes. Most of the twenty-seven transcription factors were correlated with immune infiltrating cells, immune response gene sets and immune response pathways. Furthermore, Adipocyte enhancer binding protein 1 (AEBP1), Homeobox B6 (HOXB6), Kruppel Like Factor 2 (KLF2) and RAR Related Orphan Receptor B (RORB) were selected out from twenty-seven transcription factors. ROC analysis showed that the four genes had a high diagnostic value for endometriosis. In addition, KLF2 and HOXB6 were found to play particularly important roles in multiple modules (String, WGCNA, SVM-RFE, random forest) on the gene interaction network. Using the ceRNA network, we found that NEAT1 may regulate the expressions of AEBP1, HOXB6 and RORB, while X Inactive Specific Transcript (XIST) may control the expressions of HOXB6, RORB and KLF2. Finally, we found that goserelin and dienogest may be potential drugs to regulate AEBP1, HOXB6, KLF2 and RORB through molecular docking. Conclusions AEBP1, HOXB6, KLF2, and RORB may be potential biomarkers for endometriosis. Two of them, KLF2 and HOXB6, are critical molecules in the gene interaction network of endometriosis. Discovered by molecular docking, AEBP1, HOXB6, KLF2, and RORB are targets for goserelin and dienogest.
Collapse
Affiliation(s)
- Rong Geng
- Department of Gynecology, Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, China
- Department of gynecology, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Xiaobin Huang
- Department of Gynecology, Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, China
- Department of gynecology, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Linxi Li
- Department of Gynecology, Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, China
- Department of gynecology, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Xin Guo
- Department of Gynecology, Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, China
- Department of gynecology, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Qingru Wang
- Department of Gynecology, Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, China
- Department of gynecology, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yuhua Zheng
- Department of Gynecology, Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, China
- Department of gynecology, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Xiaoling Guo
- Department of Gynecology, Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, China
- Department of gynecology, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
4
|
Nobari ST, Nojadeh JN, Talebi M. B-cell maturation antigen targeting strategies in multiple myeloma treatment, advantages and disadvantages. J Transl Med 2022; 20:82. [PMID: 35144648 PMCID: PMC8832753 DOI: 10.1186/s12967-022-03285-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 01/29/2022] [Indexed: 01/02/2023] Open
Abstract
B cell maturation antigen (BCMA), a transmembrane glycoprotein member of the tumor necrosis factor receptor superfamily 17 (TNFRSF17), highly expressed on the plasma cells of Multiple myeloma (MM) patients, as well as the normal population. BCMA is used as a biomarker for MM. Two members of the TNF superfamily proteins, including B-cell activating factor (BAFF) and A proliferation-inducing ligand (APRIL), are closely related to BCMA and play an important role in plasma cell survival and progression of MM. Despite the maximum specificity of the monoclonal antibody technologies, introducing the tumor-specific antigen(s) is not applicable for all malignancies, such as MM that there plenty of relatively specific antigens such as GPCR5D, MUC1, SLAMF7 and etc., but higher expression of BCMA on these cells in comparison with normal ones can be regarded as a relatively exclusive marker. Currently, different monoclonal antibody (mAb) technologies applied in anti-MM therapies such as daratuzumab, SAR650984, GSK2857916, and CAR-T cell therapies are some of these tools that are reviewed in the present manuscript. By the way, the structure, function, and signaling of the BCMA and related molecule(s) role in normal plasma cells and MM development, evaluated as well as the potential side effects of its targeting by different CAR-T cells generations. In conclusion, BCMA can be regarded as an ideal molecule to be targeted in immunotherapeutic methods, regarding lower potential systemic and local side effects.
Collapse
Affiliation(s)
- Shirin Teymouri Nobari
- Department of Medical Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Jafar Nouri Nojadeh
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Talebi
- Department of Applied Cells Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
5
|
Zhao J, Jiang L, Uehara M, Banouni N, Al Dulaijan BS, Azzi J, Ichimura T, Li X, Jarolim P, Fiorina P, Tullius SG, Madsen JC, Kasinath V, Abdi R. ACTH treatment promotes murine cardiac allograft acceptance. JCI Insight 2021; 6:e143385. [PMID: 34236047 PMCID: PMC8410061 DOI: 10.1172/jci.insight.143385] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 06/02/2021] [Indexed: 12/13/2022] Open
Abstract
Heart transplantation is the optimal therapy for patients with end-stage heart disease, but its long-term outcome remains inadequate. Recent studies have highlighted the importance of the melanocortin receptors (MCRs) in inflammation, but how MCRs regulate the balance between alloreactive T cells and Tregs, and whether they impact chronic heart transplant rejection, is unknown. Here, we found that Tregs express MC2R, and MC2R expression was highest among all MCRs by Tregs. Our data indicate that adrenocorticotropic hormone (ACTH), the sole ligand for MC2R, promoted the formation of Tregs by increasing the expression of IL-2Rα (CD25) in CD4+ T cells and activation of STAT5 in CD4+CD25+ T cells. ACTH treatment also improved the survival of heart allografts and increased the formation of Tregs in CD28KO mice. ACTH treatment synergized with the tolerogenic effect of CTLA-4–Ig, resulting in long-term survival of heart allografts and an increase in intragraft Tregs. ACTH administration also demonstrated higher prolongation of heart allograft survival in transgenic mouse recipients with both complete KO and conditional KO of PI3Kγ in T cells. Finally, ACTH treatment reduced chronic rejection markedly. These data demonstrate that ACTH treatment improved heart transplant outcomes, and this effect correlated with an increase in Tregs.
Collapse
Affiliation(s)
- Jing Zhao
- Transplantation Research Center.,Renal Division, and
| | - Liwei Jiang
- Transplantation Research Center.,Renal Division, and
| | - Mayuko Uehara
- Transplantation Research Center.,Renal Division, and
| | - Naima Banouni
- Transplantation Research Center.,Renal Division, and
| | | | - Jamil Azzi
- Transplantation Research Center.,Renal Division, and
| | | | - Xiaofei Li
- Transplantation Research Center.,Renal Division, and
| | - Petr Jarolim
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Paolo Fiorina
- Department of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,International Center for Type 1 Diabetes, Centro di Ricerca Pediatrica Romeo ed Enrica Invernizzi, Dipartimento di Scienze Biomediche e Cliniche "L. Sacco", Università di Milano, Milan, Italy.,Endocrinology Division, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Stefan G Tullius
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Joren C Madsen
- Center for Transplantation Sciences, Department of Surgery, and.,Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Reza Abdi
- Transplantation Research Center.,Renal Division, and
| |
Collapse
|
6
|
Huang J, Nong X, Chen Y, Zhang A, Chen L. 3-O-trans-caffeoyloleanolic acid improves acute lung injury via anti-inflammation and antioxidative stress-involved PI3K/AKT pathway. Chem Biol Drug Des 2021; 98:114-126. [PMID: 33961336 DOI: 10.1111/cbdd.13856] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 01/28/2021] [Indexed: 12/20/2022]
Abstract
3-O-trans-caffeoyloleanolic acid (COA) is a pentacyclic triterpenoid compound, with significant anti-inflammatory effects. In this study, we report the protective effects of COA on lipopolysaccharide (LPS)-induced acute lung injury (ALI) and explored its mechanism of action. LPS was used to construct in vivo mouse ALI models to observe the effects of COA pretreatment on lung pathology, inflammation, and oxidative stress. In vitro, mouse alveolar macrophages MH-S cells were cultured and stimulated with LPS to investigate the effects of COA pretreatment on inflammation and oxidative stress. Western blotting was used to investigate the expression of iNOS, TLR4, p-p65, p-AKT, and p-PI3K from in vivo and in vitro samples. The results showed that COA significantly improved lung injury, inhibited neutrophil infiltration, prevented macrophage infiltration, inhibited the release of inflammatory factors, reduced oxidative stress, and down-regulated the expression of iNOS, TLR4, p-p65, p-AKT, and p-PI3K in ALI mice caused by LPS. In vitro, COA inhibited the release of inflammatory factors, reduced oxidative stress, and down-regulated the expression of iNOS, TLR4, p-p65, p-AKT, and p-PI3K in MH-S cells stimulated with LPS. Of interest, the protective effects of COA were significantly attenuated in MH-S cells pretreated with the PI3K phosphopeptide activator 740Y-P with no effect on TLR4 expression observed. Taken together, these findings confirm the protective effects of COA on ALI. We further demonstrate that the anti-inflammation and antioxidant effects of COA are mediated through its effects on PI3K/AKT and potentially TLR4.
Collapse
Affiliation(s)
- Jianhua Huang
- Department of Respiratory and Critical Care Medicine, Jiangxi Chest (Third people) Hospital, Nanchang, China
| | - Xueping Nong
- Pathology Department, Jiangxi Chest (Third people) Hospital, Nanchang, China
| | - Yanling Chen
- Department of Respiratory and Critical Care Medicine, Jiangxi Chest (Third people) Hospital, Nanchang, China
| | - Aimei Zhang
- Department of Respiratory and Critical Care Medicine, Jiangxi Chest (Third people) Hospital, Nanchang, China
| | - Lerong Chen
- Department of Respiratory and Critical Care Medicine, Jiangxi Chest (Third people) Hospital, Nanchang, China
| |
Collapse
|
7
|
Rohrbacher L, Brauchle B, Ogrinc Wagner A, von Bergwelt-Baildon M, Bücklein VL, Subklewe M. The PI3K∂-Selective Inhibitor Idelalisib Induces T- and NK-Cell Dysfunction Independently of B-Cell Malignancy-Associated Immunosuppression. Front Immunol 2021; 12:608625. [PMID: 33790890 PMCID: PMC8005712 DOI: 10.3389/fimmu.2021.608625] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 02/11/2021] [Indexed: 11/23/2022] Open
Abstract
B-cell receptors, multiple receptor tyrosine kinases, and downstream effectors are constitutively active in chronic lymphocytic leukemia (CLL) B cells. Activation of these pathways results in resistance to apoptosis and enhanced survival of the leukemic cells. Idelalisib is a highly selective inhibitor of the PI3K p110∂ isoform and is approved for the treatment of CLL in patients with relapsed/refractory disease or in those harboring 17p deletions or tp53 mutations. Despite the initial excitement centered around high response rates in clinical trials of idelalisib, its therapeutic success has been hindered by the incidence of severe opportunistic infections. To examine the potential contribution of idelalisib to the increased risk of infection, we investigated the effects of idelalisib on the immune cell compartments of healthy donors (HDs) and CLL patients. PI3K∂ blockade by idelalisib reduced the expression levels of inhibitory checkpoint molecules in T cells isolated from both HDs and CLL patients. In addition, the presence of idelalisib in cultures significantly decreased T-cell-mediated cytotoxicity and granzyme B secretion, as well as cytokine secretion levels in both cohorts. Furthermore, idelalisib reduced the proliferation and cytotoxicity of HD NK cells. Collectively, our data demonstrate that both human T and NK cells are highly sensitive to PI3K∂ inhibition. Idelalisib interfered with the functions of T and NK cell cells from both HDs and CLL patients. Therefore, idelalisib might contribute to an increased risk of infections regardless of the underlying B-cell malignancy.
Collapse
Affiliation(s)
- Lisa Rohrbacher
- Laboratory for Translational Cancer Immunology, Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany.,Department of Internal Medicine III, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Bettina Brauchle
- Laboratory for Translational Cancer Immunology, Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany.,Department of Internal Medicine III, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Ana Ogrinc Wagner
- Laboratory for Translational Cancer Immunology, Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany.,Department of Internal Medicine III, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Michael von Bergwelt-Baildon
- Laboratory for Translational Cancer Immunology, Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany.,Department of Internal Medicine III, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany.,German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Veit L Bücklein
- Laboratory for Translational Cancer Immunology, Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany.,Department of Internal Medicine III, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Marion Subklewe
- Laboratory for Translational Cancer Immunology, Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany.,Department of Internal Medicine III, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany.,German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
| |
Collapse
|
8
|
Studying Sjögren's syndrome in mice: What is the best available model? J Oral Biol Craniofac Res 2021; 11:245-255. [PMID: 33665074 DOI: 10.1016/j.jobcr.2020.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/04/2020] [Accepted: 12/05/2020] [Indexed: 01/18/2023] Open
Abstract
Sjögren's syndrome (SS) is a common autoimmune disease characterized by lymphocytic infiltration and destruction of exocrine glands. The disease manifests primarily in the salivary and lacrimal glands, but other organs are also involved, leading to dry mouth, dry eyes, and other extra-glandular manifestations. Studying the disease in humans is entailed with many limitations and restrictions; therefore, the need for a proper mouse model is mandatory. SS mouse models are categorized, depending on the disease emergence into spontaneous or experimentally manipulated models. The usefulness of each mouse model varies depending on the SS features exhibited by that model; each SS model has advanced our understanding of the disease pathogenesis. In this review article, we list all the available murine models which have been used to study SS and we comment on the characteristics exhibited by each mouse model to assist scientists to select the appropriate model for their specific studies. We also recommend a murine strain that is the most relevant to the ideal SS model, based on our experience acquired during previous and current investigations.
Collapse
|
9
|
Avenanthramide C Prevents Neuronal Apoptosis via PI3K/Akt/GSK3β Signaling Pathway Following Middle Cerebral Artery Occlusion. Brain Sci 2020; 10:brainsci10110878. [PMID: 33233587 PMCID: PMC7699697 DOI: 10.3390/brainsci10110878] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 12/28/2022] Open
Abstract
Avenanthramides are a group of phenolic alkaloids that have been shown to have anti-inflammatory, anti-oxidant, anti-atherogenic, and vasodilation effects. The aim of the present study was to investigate the neuroprotective effect of avenanthramide-c (Avn-c) in focal brain ischemia and reperfusion injury using middle cerebral artery occlusion (MCAo) model with mice. Male C57BL/6 mice were divided into 4 groups: sham, control (MCAo), Avn-c, and Avn-c + LY294002 (phosphoinositide 3-kinase inhibitor) group. They were subjected to 60 min MCAo followed by reperfusion. Brain infarct volume and neurological deficit scores were measured after 24 h of reperfusion. We evaluated the blood brain barrier (BBB) integrity (ZO-1, VE-cadherin and occludin) and apoptosis (Bax, Bcl2, caspase3, Cytochrome C, and poly ADP ribose polymerase(PARP)-1). We also measured GSK3β for evaluation of the downstream mechanism of Akt. We examined the effect of the Avn-c in the phosphoinositide 3-kinase pathway. Avn-c reduced neurological score and infarction size. Avn-c inhibited the MCAo-induced disruption of tight junction proteins. Avn-c decreased apoptotic protein expression (Bax, Cytochrome C, and cleaved PARP-1) and increased anti-apoptotic protein expression (Bcl2) after MCAo. Akt and GSK3β were decreased in MCAo group and were restored in Avn-c group. This effect of Avn-c was abolished by PI3K inhibitor. In summary, Avn-c showed neuroprotective effects through PI3K-Akt-GSK3β signaling pathway.
Collapse
|
10
|
Capillarisin protects SH-SY5Y cells against bupivacaine-induced apoptosis via ROS-mediated PI3K/PKB pathway. Life Sci 2020; 259:118279. [DOI: 10.1016/j.lfs.2020.118279] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/05/2020] [Accepted: 08/10/2020] [Indexed: 01/10/2023]
|
11
|
An K, Qin Q, Yu S, Xue M, Wang Z, Lin Q, Ma Y, Yan G, Mo S, Chen Y, Zhang L, Zhong J, Qi Z, Xia J. Combination of N, N'-dicyclohexyl-N-arachidonic acylurea and tacrolimus prolongs cardiac allograft survival in mice. Immunol Cell Biol 2020; 98:382-396. [PMID: 32162358 DOI: 10.1111/imcb.12327] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 01/29/2020] [Accepted: 03/09/2020] [Indexed: 12/27/2022]
Abstract
Current immunosuppressive agents for organ transplantation are not ideal because of their strong toxicity and adverse effects. Hence, there is an urgent need to develop novel immunosuppressive agents. The compound N, N'-dicyclohexyl-N-arachidonic acylurea (DCAAA) is a novel highly unsaturated fatty acid from the traditional Chinese medicinal plant Radix Isatidis. In this study, we systematically investigated the toxicity, immunosuppressive effect and mechanisms underlying the activity of DCAAA. The toxicity tests showed that DCAAA treatment did not lead to red blood cell hemolysis and did not affect the liver and kidney functions in mice. The lymphocyte transformation test showed that DCAAA treatment inhibited lymphocyte proliferation in a dose-dependent manner. An in vivo cardiac allotransplantation experiment showed that DCAAA treatment could suppress the immune rejection and significantly prolong the survival of cardiac allografts in recipient mice by reducing the proportion of CD4+ T cells in the spleen and grafts, concentration of interferon-γ in the supernatant and serum and infiltration of inflammatory cells into the grafts. Moreover, a combination treatment with DCAAA and tacrolimus had a synergistic effect in preventing acute rejection of heart transplants. In vitro molecular biology experiments showed that DCAAA treatment inhibited activation of the T-cell receptor-mediated phosphoinostide 3-kinase-protein kinase B pathway, thereby arresting cell cycle transition from the G1 to the S phase, and inhibiting lymphocyte proliferation. Overall, our study reveals a novel, low-toxicity immunosuppressive agent that has the potential to reduce the toxic side effects of existing immunosuppressive agents when used in combination with them.
Collapse
Affiliation(s)
- Ke An
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Qing Qin
- Department of Natural Product Chemistry, School of Pharmaceutical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Shengnan Yu
- Department of Obstetrics and Gynecology, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, China
| | - Mengjiao Xue
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Zhenzhen Wang
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Qingru Lin
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yunhan Ma
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Guoliang Yan
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Sirui Mo
- Youjiang Medical University For Nationalities, Baise, Guangxi, China
| | - Yingyu Chen
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Liyi Zhang
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Jiaying Zhong
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Zhongquan Qi
- School of Medicine, Guangxi University, Nanning, Guangxi, China
| | - Junjie Xia
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
12
|
Zhang H, Liu X, Zhang X, Qin N, Xu K, Yin W, Zheng Y, Song Y, Zeng R, Liu J. Phosphoinositide 3-Kinase Promotes Oxidative Burst, Stomatal Closure and Plant Immunity in Bacterial Invasion. FRONTIERS IN PLANT SCIENCE 2020; 10:1740. [PMID: 32117334 PMCID: PMC7025545 DOI: 10.3389/fpls.2019.01740] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 12/11/2019] [Indexed: 05/27/2023]
Abstract
Phosphoinositide 3-kinase (PI3K) plays a vital role in plant response to abiotic stress. However, the role of PI3K in plant immunity is largely unknown. This study showed that PI3K enhanced Arabidopsis resistance to Pseudomonas syringae pv tomato DC3000 (Pst DC3000) and Pst DC3000 (avrRpt2). Overexpression of AtVPS34 promoted stomatal closure while PI3K inhibitors blocked that after spray inoculation. Additionally, gene expression of AtVPS34 was increased upon infection by Pst DC3000 (avrRpt2), and SA upregulated AtVPS34 gene expression in this process. Furthermore, overexpression of AtVPS34 enhanced PR gene expression after syringe infiltration with Pst DC3000 (avrRpt2), while PI3K inhibitors inhibited that. The production of hydrogen peroxide and the expression of gene encoding antioxidant enzyme were both enhanced in AtVPS34 overexpressing lines after spray inoculation or syringe infiltration with Pst DC3000 (avrRpt2). Collectively, these results unraveled a novel and broad role of PI3K in plant immunity which promoted stomatal closure and PR gene expression possibly via regulating ROS production.
Collapse
Affiliation(s)
- Huiying Zhang
- Fujian Provincial Key Laboratory of Plant Functional Biology, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xin Liu
- Fujian Provincial Key Laboratory of Plant Functional Biology, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xiyong Zhang
- Fujian Provincial Key Laboratory of Plant Functional Biology, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Ningning Qin
- Fujian Provincial Key Laboratory of Plant Functional Biology, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Kaifang Xu
- Key Laboratory of Ministry of Education for Genetics, Breeding and Multiple Utilization of Crops, College of Agriculture, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Weihua Yin
- Fujian Provincial Key Laboratory of Plant Functional Biology, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Yueqin Zheng
- Fujian Provincial Key Laboratory of Plant Functional Biology, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Yuanyuan Song
- Key Laboratory of Ministry of Education for Genetics, Breeding and Multiple Utilization of Crops, College of Agriculture, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Rensen Zeng
- Key Laboratory of Ministry of Education for Genetics, Breeding and Multiple Utilization of Crops, College of Agriculture, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Jian Liu
- Key Laboratory of Ministry of Education for Genetics, Breeding and Multiple Utilization of Crops, College of Agriculture, Fujian Agriculture and Forestry University, Fuzhou, China
| |
Collapse
|
13
|
Yousefi Z, Sharifzadeh S, Yar-Ahmadi V, Andalib A, Eskandari N. Fc Receptor-Like 1 as a Promising Target for Immunotherapeutic Interventions of B-Cell-Related Disorders. Biomark Insights 2019; 14:1177271919882351. [PMID: 31798301 PMCID: PMC6864034 DOI: 10.1177/1177271919882351] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 09/22/2019] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Human B-cell responses are regulated through synergy between a collection of activation and inhibitory receptors. Fc receptor-like (FCRL) molecules have recently been identified as co-receptors that are preferentially expressed in human B-cells, which may also play an important role in the regulation of human B-cell responses. FCRL1 is a member of the FCRL family molecules with 2 immunoreceptor tyrosine-based activation motifs (ITAMs) in its cytoplasmic tail. This study aimed to investigate the regulatory roles of FCRL1 in human B-cell responses. MATERIALS AND METHODS The regulatory potential of FCRL1 in human B-cell through knockdown of FCRL1 expression in the Ramos and Daudi Burkitt lymphoma (BL) cell lines by using the retroviral-based short hairpin ribonucleic acid (shRNA) delivery method. The functional consequences of FCRL1 knockdown were assessed by measuring the proliferation, apoptosis, and the expression levels of Bcl-2, Bid, and Bax genes as well as phosphoinositide-3 kinase/-serine-threonine kinase AKT (PI3K/p-AKT) pathway in the BL cells, using the quantitative real-time polymerase chain reaction (PCR) and flow cytometry analysis. The NF-κB activity was also measured by the enzyme-linked immunosorbent assay (ELISA). RESULTS FCRL1 knockdown significantly decreased cell proliferation and increased apoptotic cell death in the BL cells. There was a significant reduction in the extent of the Bcl-2 gene expression in the treated BL cells compared with control cells. On the contrary, FCRL1 knockdown increased the expression levels of Bid and Bax genes in the treated BL cells when compared with control cells. In addition, the extent of the PI3K/p-AKT expression and phosphorylated-p65 NF-κB activity was significantly decreased in the treated BL cells compared with control cells. CONCLUSIONS These results suggest that FCRL1 can play a key role in the activation of human B-cell responses and has the potential to serve as a target for immunotherapy of FCRL1 positive B-cell-related disorders.
Collapse
Affiliation(s)
- Zahra Yousefi
- Department of Immunology, Faculty of
Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sedigheh Sharifzadeh
- Diagnostic Laboratory Sciences and
Technology Research Center, School of Paramedical Sciences, Shiraz University of
Medical Sciences, Shiraz, Iran
| | - Vali Yar-Ahmadi
- Department of Parasitology, Faculty of
Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Alireza Andalib
- Department of Immunology, Faculty of
Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nahid Eskandari
- Department of Immunology, Faculty of
Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Applied Physiology Research Center,
Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
14
|
Peptide Tk-PQ induces immunosuppression in skin allogeneic transplantation via increasing Foxp3 + Treg and impeding nuclear translocation of NF-κB. Mol Immunol 2018; 101:597-607. [PMID: 30001873 DOI: 10.1016/j.molimm.2018.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 05/09/2018] [Accepted: 05/09/2018] [Indexed: 12/14/2022]
Abstract
Solid organ transplantation is used as the last resort for patients with end-stage disease, but allograft rejection is an unsolved problem. Here, we showed that Tk-PQ, a peptide derived from trichosanthin, had an immune-suppressive effect without obvious cytotoxicity in vitro and in a mouse skin allo-transplantation model. In vitro, treatment of Tk-PQ administrated type 2 T helper cell (Th2)/regulatory T-cell (Treg) cytokines, and increased the ratio of CD4+CD25+Foxp3+ Treg by repressing the PI3K/mTOR pathway. In addition, Tk-PQ decreased NF-κB activation to downregulate pro-inflammatory cytokines. Tk-PQ treatment in the mouse skin transplantation model also caused the similar molecular and cellular phenotypes. Furthermore, Tk-PQ enhanced the suppressive function of Treg by increasing Foxp3 expression, and substantially improved allograft survival. These finding demonstrate that Tk-PQ has the potential to be used in clinical allogeneic transplantation.
Collapse
|
15
|
Ben Hamda C, Sangeda R, Mwita L, Meintjes A, Nkya S, Panji S, Mulder N, Guizani-Tabbane L, Benkahla A, Makani J, Ghedira K, H3ABioNet Consortium. A common molecular signature of patients with sickle cell disease revealed by microarray meta-analysis and a genome-wide association study. PLoS One 2018; 13:e0199461. [PMID: 29979707 PMCID: PMC6034806 DOI: 10.1371/journal.pone.0199461] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 06/07/2018] [Indexed: 12/16/2022] Open
Abstract
A chronic inflammatory state to a large extent explains sickle cell disease (SCD) pathophysiology. Nonetheless, the principal dysregulated factors affecting this major pathway and their mechanisms of action still have to be fully identified and elucidated. Integrating gene expression and genome-wide association study (GWAS) data analysis represents a novel approach to refining the identification of key mediators and functions in complex diseases. Here, we performed gene expression meta-analysis of five independent publicly available microarray datasets related to homozygous SS patients with SCD to identify a consensus SCD transcriptomic profile. The meta-analysis conducted using the MetaDE R package based on combining p values (maxP approach) identified 335 differentially expressed genes (DEGs; 224 upregulated and 111 downregulated). Functional gene set enrichment revealed the importance of several metabolic pathways, of innate immune responses, erythrocyte development, and hemostasis pathways. Advanced analyses of GWAS data generated within the framework of this study by means of the atSNP R package and SIFT tool identified 60 regulatory single-nucleotide polymorphisms (rSNPs) occurring in the promoter of 20 DEGs and a deleterious SNP, affecting CAMKK2 protein function. This novel database of candidate genes, transcription factors, and rSNPs associated with SCD provides new markers that may help to identify new therapeutic targets.
Collapse
Affiliation(s)
- Cherif Ben Hamda
- Laboratory of Bioinformatics, Biomathematics and Biostatistics, Institute Pasteur of Tunis, Tunis, Tunisia
- University of Tunis El Manar, Tunis, Tunisia
- Faculty of Science of Bizerte, Jarzouna, University of Carthage, Tunisia
- * E-mail: (KG); (CBH)
| | - Raphael Sangeda
- Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Liberata Mwita
- Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | | | - Siana Nkya
- Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Sumir Panji
- University of Cape Town, Cape Town, South Africa
| | | | - Lamia Guizani-Tabbane
- University of Tunis El Manar, Tunis, Tunisia
- Laboratory of Medical Parasitology, Biotechnology and Biomolecules, Institute Pasteur of Tunis, Tunis, Tunisia
| | - Alia Benkahla
- Laboratory of Bioinformatics, Biomathematics and Biostatistics, Institute Pasteur of Tunis, Tunis, Tunisia
- University of Tunis El Manar, Tunis, Tunisia
| | - Julie Makani
- Faculty of Science of Bizerte, Jarzouna, University of Carthage, Tunisia
| | - Kais Ghedira
- Laboratory of Bioinformatics, Biomathematics and Biostatistics, Institute Pasteur of Tunis, Tunis, Tunisia
- University of Tunis El Manar, Tunis, Tunisia
- * E-mail: (KG); (CBH)
| | | |
Collapse
|
16
|
Wang X, Ha T, Hu Y, Lu C, Liu L, Zhang X, Kao R, Kalbfleisch J, Williams D, Li C. MicroRNA-214 protects against hypoxia/reoxygenation induced cell damage and myocardial ischemia/reperfusion injury via suppression of PTEN and Bim1 expression. Oncotarget 2018; 7:86926-86936. [PMID: 27894079 PMCID: PMC5349964 DOI: 10.18632/oncotarget.13494] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 10/28/2016] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Myocardial apoptosis plays an important role in myocardial ischemia/reperfusion (I/R) injury. Activation of PI3K/Akt signaling protects the myocardium from I/R injury. This study investigated the role of miR-214 in hypoxia/reoxygenation (H/R)-induced cell damage in vitro and myocardial I/R injury in vivo. METHODS AND RESULTS H9C2 cardiomyoblasts were transfected with lentivirus expressing miR-214 (LmiR-214) or lentivirus expressing scrambled miR-control (LmiR-control) respectively, to establish cell lines of LmiR-214 and LmiR-control. The cells were subjected to hypoxia for 4 h followed by reoxygenation for 24 h. Transfection of LmiR-214 suppresses PTEN expression, significantly increases the levels of Akt phosphorylation, markedly attenuates LDH release, and enhances the viability of the cells subjected to H/R. In vivo transfection of mouse hearts with LmiR-214 significantly attenuates I/R induced cardiac dysfunction and reduces I/R-induced myocardial infarct size. LmiR-214 transfection significantly attenuates I/R-induced myocardial apoptosis and caspase-3/7 and caspase-8 activity. Increased expression of miR-214 by transfection of LmiR-214 suppresses PTEN expression, increases the levels of phosphorylated Akt, represses Bim1 expression and induces Bad phosphorylation in the myocardium. In addition, in vitro data shows transfection of miR-214 mimics to H9C2 cells suppresses the expression and translocation of Bim1 from cytosol to mitochondria and induces Bad phosphorylation. CONCLUSIONS Our in vitro and in vivo data suggests that miR-214 protects cells from H/R induced damage and attenuates I/R induced myocardial injury. The mechanisms involve activation of PI3K/Akt signaling by targeting PTEN expression, induction of Bad phosphorylation, and suppression of Bim1 expression, resulting in decreases in I/R-induced myocardial apoptosis.
Collapse
Affiliation(s)
- Xiaohui Wang
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Tuanzhu Ha
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA.,Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Yuanping Hu
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Chen Lu
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Li Liu
- Department of Geriatrics, First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Xia Zhang
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Race Kao
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA.,Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - John Kalbfleisch
- Department of Biometry and Medical Computing, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA.,Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - David Williams
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA.,Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Chuanfu Li
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA.,Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| |
Collapse
|
17
|
Wogonoside prevents colitis-associated colorectal carcinogenesis and colon cancer progression in inflammation-related microenvironment via inhibiting NF-κB activation through PI3K/Akt pathway. Oncotarget 2018; 7:34300-15. [PMID: 27102438 PMCID: PMC5085157 DOI: 10.18632/oncotarget.8815] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 03/28/2016] [Indexed: 12/31/2022] Open
Abstract
The inflammatory microenvironment has been reported to be correlated with tumor initiation and malignant development. In the previous studies we have found that wogonoside exerts anti-neoplastic and anti-inflammatory activities. In this study, we aimed to further investigate the chemopreventive effects of wogonoside on colitis-associated cancer and delineated the potential mechanisms. In the azoxymethane initiated and dextran sulfate sodium (AOM/DSS) promoted colorectal carcinogenesis mouse model, wogonoside significantly reduced the disease severity, lowered tumor incidence and inhibited the development of colorectal adenomas. Moreover, wogonoside inhibited inflammatory cells infiltration and cancer cell proliferation at tumor site. Furthermore, wogonoside dramatically decreased the secretion and expression of IL-1β, IL-6 and TNF-α as well as the nuclear expression of NF-κB in adenomas and surrounding tissues. In vitro results showed that wogonoside suppressed the proliferation of human colon cancer cells in the inflammatory microenvironment. Mechanistically, we found that wogonoside inhibited NF-κB activation via PI3K/Akt pathway. In conclusion, our results demonstrated that wogonoside attenuated colitis-associated tumorigenesis in mice and inhibited the progression of human colon cancer in inflammation-related microenvironment via suppressing NF-κB activation by PI3K/Akt pathway, indicating that wogonoside could be a promising therapeutic agent for colorectal cancer.
Collapse
|
18
|
Dobranowski P, Sly LM. SHIP negatively regulates type II immune responses in mast cells and macrophages. J Leukoc Biol 2018; 103:1053-1064. [PMID: 29345374 DOI: 10.1002/jlb.3mir0817-340r] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 12/11/2017] [Accepted: 12/14/2017] [Indexed: 12/13/2022] Open
Abstract
SHIP is a hematopoietic-specific lipid phosphatase that dephosphorylates PI3K-generated PI(3,4,5)-trisphosphate. SHIP removes this second messenger from the cell membrane blunting PI3K activity in immune cells. Thus, SHIP negatively regulates mast cell activation downstream of multiple receptors. SHIP has been referred to as the "gatekeeper" of mast cell degranulation as loss of SHIP dramatically increases degranulation or permits degranulation in response to normally inert stimuli. SHIP also negatively regulates Mϕ activation, including both pro-inflammatory cytokine production downstream of pattern recognition receptors, and alternative Mϕ activation by the type II cytokines, IL-4, and IL-13. In the SHIP-deficient (SHIP-/- ) mouse, increased mast cell and Mϕ activation leads to spontaneous inflammatory pathology at mucosal sites, which is characterized by high levels of type II inflammatory cytokines. SHIP-/- mast cells and Mϕs have both been implicated in driving inflammation in the SHIP-/- mouse lung. SHIP-/- Mϕs drive Crohn's disease-like intestinal inflammation and fibrosis, which is dependent on heightened responses to innate immune stimuli generating IL-1, and IL-4 inducing abundant arginase I. Both lung and gut pathology translate to human disease as low SHIP levels and activity have been associated with allergy and with Crohn's disease in people. In this review, we summarize seminal literature and recent advances that provide insight into SHIP's role in mast cells and Mϕs, the contribution of these cell types to pathology in the SHIP-/- mouse, and describe how these findings translate to human disease and potential therapies.
Collapse
Affiliation(s)
- Peter Dobranowski
- Division of Gastroenterology, Department of Pediatrics, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Laura M Sly
- Division of Gastroenterology, Department of Pediatrics, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
19
|
Uehara M, McGrath MM, Ohori S, Solhjou Z, Banouni N, Routray S, Evans C, DiNitto JP, Elkhal A, Turka LA, Strom TB, Tullius SG, Winkler DG, Azzi J, Abdi R. Regulation of T cell alloimmunity by PI3Kγ and PI3Kδ. Nat Commun 2017; 8:951. [PMID: 29038423 PMCID: PMC5643371 DOI: 10.1038/s41467-017-00982-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 08/10/2017] [Indexed: 01/04/2023] Open
Abstract
Phosphatidylinositol-3-kinases (PI3K) γ and δ are preferentially enriched in leukocytes, and defects in these signaling pathways have been shown to impair T cell activation. The effects of PI3Kγ and PI3Kδ on alloimmunity remain underexplored. Here, we show that both PI3Kγ−/− and PI3KδD910A/D910A mice receiving heart allografts have suppression of alloreactive T effector cells and delayed acute rejection. However, PI3Kδ mutation also dampens regulatory T cells (Treg). After treatment with low dose CTLA4-Ig, PI3Kγ−/−, but not PI3ΚδD910A/D910A, recipients exhibit indefinite prolongation of heart allograft survival. PI3KδD910A/D910A Tregs have increased apoptosis and impaired survival. Selective inhibition of PI3Kγ and PI3Kδ (using PI3Kδ and dual PI3Kγδ chemical inhibitors) shows that PI3Kγ inhibition compensates for the negative effect of PI3Kδ inhibition on long-term allograft survival. These data serve as a basis for future PI3K-based immune therapies for transplantation. Phosphatidylinositol-3-kinases (PI3K) γ and δ are key regulators of T cell signaling. Here the author show, using mouse heart allograft transplantation models, that PI3Kγ or PI3Kδ deficiency prolongs graft survival, but selective inhibition of PI3Kγ or PI3Kδ reveals alternative transplant survival outcomes post CTLA4-Ig treatment.
Collapse
Affiliation(s)
- Mayuko Uehara
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Harvard Medical School, 221 Longwood Avenue, Boston, MA, 02115, USA
| | - Martina M McGrath
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Harvard Medical School, 221 Longwood Avenue, Boston, MA, 02115, USA
| | - Shunsuke Ohori
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Harvard Medical School, 221 Longwood Avenue, Boston, MA, 02115, USA
| | - Zhabiz Solhjou
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Harvard Medical School, 221 Longwood Avenue, Boston, MA, 02115, USA
| | - Naima Banouni
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Harvard Medical School, 221 Longwood Avenue, Boston, MA, 02115, USA
| | - Sujit Routray
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Harvard Medical School, 221 Longwood Avenue, Boston, MA, 02115, USA
| | - Catherine Evans
- Infinity Pharmaceuticals, Inc 784 Memorial Drive, Cambridge, MA, 02139, USA
| | - Jonathan P DiNitto
- Infinity Pharmaceuticals, Inc 784 Memorial Drive, Cambridge, MA, 02139, USA
| | - Abdallah Elkhal
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Brigham and Women's Hospital and Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Laurence A Turka
- Center for Transplantation Sciences, Massachusetts General Hospital/Harvard Medical School, Massachusetts Massachusetts General Hospital-East Charlestown Navy Yard Building 149, 13th Street, Charlestown, MA, 02129-2020, USA
| | - Terry B Strom
- The Transplant Institute, Beth Israel Deaconess Medical Center/Harvard Medical School, 330 Brookline Avenue, E/CLS Room 607, Boston, MA, 02215, USA
| | - Stefan G Tullius
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Brigham and Women's Hospital and Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - David G Winkler
- Infinity Pharmaceuticals, Inc 784 Memorial Drive, Cambridge, MA, 02139, USA
| | - Jamil Azzi
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Harvard Medical School, 221 Longwood Avenue, Boston, MA, 02115, USA
| | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital and Harvard Medical School, 221 Longwood Avenue, Boston, MA, 02115, USA.
| |
Collapse
|
20
|
Hu L, Cai N, Jia H. Pterostilbene attenuates myocardial ischemia-reperfusion injury via the phosphatidylinositol 3'-kinase-protein kinase B signaling pathway. Exp Ther Med 2017; 14:5509-5514. [PMID: 29285084 DOI: 10.3892/etm.2017.5246] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 06/23/2017] [Indexed: 11/06/2022] Open
Abstract
The current study aimed to evaluate the cardioprotective effects of pterostilbene (PTB) on myocardial ischemia-reperfusion (I/R) injury in rats and identify its possible underlying mechanisms of action. A rat I/R model was established by ligating the left anterior descending coronary artery for 30 min and releasing the ligature to induce reperfusion for 120 min. Serum creatine kinase-MB (CK-MB) and lactate dehydrogenase (LDH) levels were measured using CK-MB and LDH assay kits and myeloperoxidase (MPO) activity in the myocardium was evaluated using an MPO assay kit. Tumor necrosis factor-α, interleukin (IL)-6 and IL-8 levels were assayed using ELISA kits. Cardiomyocyte apoptosis was measured using terminal deoxynucleotidyl transferase dUTP nick end labeling staining. Levels of protein kinase B (Akt) and phosphorylated Akt (p-Akt) were measured using western blotting. The results demonstrated that treatment with PTB significantly reduced cardiomyocyte apoptosis, significantly increased Bcl-2 and p-Akt levels and decreased Bax expression in the hearts of rats subjected to I/R injury. However, the protective effects induced by PTB were attenuated by LY294002, which inhibits Akt activation. The results of the current study suggest that PTB treatment may reduce the I/R injury-induced apoptosis of cardiomyocytes, which is mediated by the phosphoinositide 3-kinase/Akt signaling pathway.
Collapse
Affiliation(s)
- Lei Hu
- Department of Clinical Laboratory, Northwest Women and Children's Hospital, Xi'an, Shaanxi 710061, P.R. China
| | - Na Cai
- Department of Medical Genetics, Northwest Women and Children's Hospital, Xi'an, Shaanxi 710061, P.R. China
| | - Hui Jia
- Department of Clinical Laboratory, Northwest Women and Children's Hospital, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
21
|
Marcoux D, Qin LY, Ruan Z, Shi Q, Ruan Q, Weigelt C, Qiu H, Schieven G, Hynes J, Bhide R, Poss M, Tino J. Identification of highly potent and selective PI3Kδ inhibitors. Bioorg Med Chem Lett 2017; 27:2849-2853. [DOI: 10.1016/j.bmcl.2017.01.077] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 01/23/2017] [Accepted: 01/25/2017] [Indexed: 01/22/2023]
|
22
|
Liu Q, Shi Q, Marcoux D, Batt DG, Cornelius L, Qin LY, Ruan Z, Neels J, Beaudoin-Bertrand M, Srivastava AS, Li L, Cherney RJ, Gong H, Watterson SH, Weigelt C, Gillooly KM, McIntyre KW, Xie JH, Obermeier MT, Fura A, Sleczka B, Stefanski K, Fancher RM, Padmanabhan S, Rp T, Kundu I, Rajareddy K, Smith R, Hennan JK, Xing D, Fan J, Levesque PC, Ruan Q, Pitt S, Zhang R, Pedicord D, Pan J, Yarde M, Lu H, Lippy J, Goldstine C, Skala S, Rampulla RA, Mathur A, Gupta A, Arunachalam PN, Sack JS, Muckelbauer JK, Cvijic ME, Salter-Cid LM, Bhide RS, Poss MA, Hynes J, Carter PH, Macor JE, Ruepp S, Schieven GL, Tino JA. Identification of a Potent, Selective, and Efficacious Phosphatidylinositol 3-Kinase δ (PI3Kδ) Inhibitor for the Treatment of Immunological Disorders. J Med Chem 2017; 60:5193-5208. [PMID: 28541707 DOI: 10.1021/acs.jmedchem.7b00618] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PI3Kδ plays an important role controlling immune cell function and has therefore been identified as a potential target for the treatment of immunological disorders. This article highlights our work toward the identification of a potent, selective, and efficacious PI3Kδ inhibitor. Through careful SAR, the successful replacement of a polar pyrazole group by a simple chloro or trifluoromethyl group led to improved Caco-2 permeability, reduced Caco-2 efflux, reduced hERG PC activity, and increased selectivity profile while maintaining potency in the CD69 hWB assay. The optimization of the aryl substitution then identified a 4'-CN group that improved the human/rodent correlation in microsomal metabolic stability. Our lead molecule is very potent in PK/PD assays and highly efficacious in a mouse collagen-induced arthritis model.
Collapse
Affiliation(s)
- Qingjie Liu
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Qing Shi
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - David Marcoux
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Douglas G Batt
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Lyndon Cornelius
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Lan-Ying Qin
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Zheming Ruan
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - James Neels
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Myra Beaudoin-Bertrand
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Anurag S Srivastava
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Ling Li
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Robert J Cherney
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Hua Gong
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Scott H Watterson
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Carolyn Weigelt
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Kathleen M Gillooly
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Kim W McIntyre
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Jenny H Xie
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Mary T Obermeier
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Aberra Fura
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Bogdan Sleczka
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Kevin Stefanski
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - R M Fancher
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Shweta Padmanabhan
- Department of Discovery Synthesis, Biocon Bristol-Myers Squibb Research Centre , Biocon Park, Bommasandra IV Phase, Jigani Link Road, Bengaluru 560099, India
| | - Thatipamula Rp
- Department of Discovery Synthesis, Biocon Bristol-Myers Squibb Research Centre , Biocon Park, Bommasandra IV Phase, Jigani Link Road, Bengaluru 560099, India
| | - Ipsit Kundu
- Department of Discovery Synthesis, Biocon Bristol-Myers Squibb Research Centre , Biocon Park, Bommasandra IV Phase, Jigani Link Road, Bengaluru 560099, India
| | | | - Rodney Smith
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - James K Hennan
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Dezhi Xing
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Jingsong Fan
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Paul C Levesque
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Qian Ruan
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Sidney Pitt
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Rosemary Zhang
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Donna Pedicord
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Jie Pan
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Melissa Yarde
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Hao Lu
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Jonathan Lippy
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Christine Goldstine
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Stacey Skala
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Richard A Rampulla
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Arvind Mathur
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Anuradha Gupta
- Department of Discovery Synthesis, Biocon Bristol-Myers Squibb Research Centre , Biocon Park, Bommasandra IV Phase, Jigani Link Road, Bengaluru 560099, India
| | - Pirama Nayagam Arunachalam
- Department of Discovery Synthesis, Biocon Bristol-Myers Squibb Research Centre , Biocon Park, Bommasandra IV Phase, Jigani Link Road, Bengaluru 560099, India
| | - John S Sack
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Jodi K Muckelbauer
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Mary Ellen Cvijic
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Luisa M Salter-Cid
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Rajeev S Bhide
- Department of Discovery Synthesis, Biocon Bristol-Myers Squibb Research Centre , Biocon Park, Bommasandra IV Phase, Jigani Link Road, Bengaluru 560099, India
| | - Michael A Poss
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - John Hynes
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Percy H Carter
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | | | - Stefan Ruepp
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Gary L Schieven
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| | - Joseph A Tino
- Research & Development, Bristol-Myers Squibb Company , Route 206 and Province Line Road, Princeton, New Jersey 08543, United States
| |
Collapse
|
23
|
78495111110.3390/cancers9050052" />
Abstract
The epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase that is commonly upregulated in cancers such as in non-small-cell lung cancer, metastatic colorectal cancer, glioblastoma, head and neck cancer, pancreatic cancer, and breast cancer. Various mechanisms mediate the upregulation of EGFR activity, including common mutations and truncations to its extracellular domain, such as in the EGFRvIII truncations, as well as to its kinase domain, such as the L858R and T790M mutations, or the exon 19 truncation. These EGFR aberrations over-activate downstream pro-oncogenic signaling pathways, including the RAS-RAF-MEK-ERK MAPK and AKT-PI3K-mTOR pathways. These pathways then activate many biological outputs that are beneficial to cancer cell proliferation, including their chronic initiation and progression through the cell cycle. Here, we review the molecular mechanisms that regulate EGFR signal transduction, including the EGFR structure and its mutations, ligand binding and EGFR dimerization, as well as the signaling pathways that lead to G1 cell cycle progression. We focus on the induction of CYCLIN D expression, CDK4/6 activation, and the repression of cyclin-dependent kinase inhibitor proteins (CDKi) by EGFR signaling pathways. We also discuss the successes and challenges of EGFR-targeted therapies, and the potential for their use in combination with CDK4/6 inhibitors.
Collapse
|
24
|
Wee P, Wang Z. Epidermal Growth Factor Receptor Cell Proliferation Signaling Pathways. Cancers (Basel) 2017; 9:cancers9050052. [PMID: 28513565 PMCID: PMC5447962 DOI: 10.3390/cancers9050052] [Citation(s) in RCA: 1211] [Impact Index Per Article: 151.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 05/10/2017] [Accepted: 05/10/2017] [Indexed: 12/12/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase that is commonly upregulated in cancers such as in non-small-cell lung cancer, metastatic colorectal cancer, glioblastoma, head and neck cancer, pancreatic cancer, and breast cancer. Various mechanisms mediate the upregulation of EGFR activity, including common mutations and truncations to its extracellular domain, such as in the EGFRvIII truncations, as well as to its kinase domain, such as the L858R and T790M mutations, or the exon 19 truncation. These EGFR aberrations over-activate downstream pro-oncogenic signaling pathways, including the RAS-RAF-MEK-ERK MAPK and AKT-PI3K-mTOR pathways. These pathways then activate many biological outputs that are beneficial to cancer cell proliferation, including their chronic initiation and progression through the cell cycle. Here, we review the molecular mechanisms that regulate EGFR signal transduction, including the EGFR structure and its mutations, ligand binding and EGFR dimerization, as well as the signaling pathways that lead to G1 cell cycle progression. We focus on the induction of CYCLIN D expression, CDK4/6 activation, and the repression of cyclin-dependent kinase inhibitor proteins (CDKi) by EGFR signaling pathways. We also discuss the successes and challenges of EGFR-targeted therapies, and the potential for their use in combination with CDK4/6 inhibitors.
Collapse
Affiliation(s)
- Ping Wee
- Department of Medical Genetics and Signal Transduction Research Group, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| | - Zhixiang Wang
- Department of Medical Genetics and Signal Transduction Research Group, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| |
Collapse
|
25
|
Huang W, Fan W, Wang Y, Han D, Li X, Li S, Li C, Xu B, Huang Y, Fu X, Cao F. Mesenchymal stem cells in alleviating sepsis-induced mice cardiac dysfunction via inhibition of mTORC1-p70S6K signal pathway. Cell Death Discov 2017; 3:16097. [PMID: 28250969 PMCID: PMC5327616 DOI: 10.1038/cddiscovery.2016.97] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 10/14/2016] [Accepted: 10/21/2016] [Indexed: 12/30/2022] Open
Abstract
Sepsis-induced cardiac dysfunction remains a major cause of morbidity and mortality in patients suffered from severe trauma. Mesenchymal stem cells (MSCs) -based treatment has been verified as a promising approach to mitigate the sepsis-induced cardiac dysfunction, but the mechanism is still ambiguous. Thus, our study was designed to explore the potential role of MSCs in sepsis-induced cardiac dysfunction. In vivo bioluminescence imaging revealed 80% acute donor cell death of bone marrow-derived MSCs (BM-MSCs) within 3 days after transplantation. However, echocardiography demonstrated that systolic function in wild-type mice group were reduced after sepsis, while the cardiac function was relatively well persevered in cardiac-conditional deletion of Raptor (component of mTORC1 complex) mice group. Raptor KO group treated with BM-MSCs appeared better cardiac function than other groups (P<0.05). In vitro cell study revealed that co-culture of H9C2 (Raptor-Knock down) and BM-MSC could attenuate the level of proinflammatory cytokines and promote the expression of anti-inflammatory cytokine accompanied by mTORC2-Akt activation (P<0.05). In contrast, co-culture H9C2 (Raptor-O.E) and BM-MSC could aggravate the inflammatory response accompanied by the activation of mTORC1-p70S6K and inhibition of mTORC2-Akt (P<0.05). The immunomodulatory property of MSC is related to the inhibition of mTORC1-p70S6K and activation of mTORC2-Akt signaling pathway. mTORC1-p70S6K and mTORC2-Akt pathways were involved in the therapeutic adjuncts of MSC. The possible mechanism due to MSC`s immunomodulatory property through activation of mTORC2-Akt and inhibition of mTORC1-p70S6K signal pathways which may lead to modulate the expression of inflammation cytokines.
Collapse
Affiliation(s)
- Wei Huang
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China; Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Shanxi, China
| | - Wensi Fan
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China; Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Shanxi, China
| | - Yabin Wang
- Department of Cardiology, Chinese PLA General Hospital , Beijing, China
| | - Dong Han
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University , Shanxi, China
| | - Xiujuan Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University , Shanxi, China
| | - Shuang Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University , Shanxi, China
| | - Congye Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University , Shanxi, China
| | - Bin Xu
- Department of Cardiology, Chinese PLA General Hospital , Beijing, China
| | - Yuesheng Huang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, The Third Military Medical University , Chongqing, China
| | - Xiaobin Fu
- Department of Cardiology, Chinese PLA General Hospital , Beijing, China
| | - Feng Cao
- Department of Cardiology, Chinese PLA General Hospital , Beijing, China
| |
Collapse
|
26
|
Ren D, Lin D, Alim A, Zheng Q, Yang X. Chemical characterization of a novel polysaccharide ASKP-1 from Artemisia sphaerocephala Krasch seed and its macrophage activation via MAPK, PI3k/Akt and NF-κB signaling pathways in RAW264.7 cells. Food Funct 2017; 8:1299-1312. [DOI: 10.1039/c6fo01699e] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The aim of this study was to investigate the molecular mechanism underlying the immunomodulatory effect of the purified Artemisia sphaerocephala Krasch seed polysaccharide (ASKP-1) in RAW264.7 macrophages.
Collapse
Affiliation(s)
- Daoyuan Ren
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control
- College of Food Engineering and Nutritional Science
- Shaanxi Normal University
- Xi'an 710062
- China
| | - Dehui Lin
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control
- College of Food Engineering and Nutritional Science
- Shaanxi Normal University
- Xi'an 710062
- China
| | - Aamina Alim
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control
- College of Food Engineering and Nutritional Science
- Shaanxi Normal University
- Xi'an 710062
- China
| | - Quan Zheng
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control
- College of Food Engineering and Nutritional Science
- Shaanxi Normal University
- Xi'an 710062
- China
| | - Xingbin Yang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control
- College of Food Engineering and Nutritional Science
- Shaanxi Normal University
- Xi'an 710062
- China
| |
Collapse
|
27
|
Chen PH, Yao H, Huang LJS. Cytokine Receptor Endocytosis: New Kinase Activity-Dependent and -Independent Roles of PI3K. Front Endocrinol (Lausanne) 2017; 8:78. [PMID: 28507533 PMCID: PMC5410625 DOI: 10.3389/fendo.2017.00078] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 03/30/2017] [Indexed: 12/14/2022] Open
Abstract
Type I and II cytokine receptors are cell surface sensors that bind cytokines in the extracellular environment and initiate intracellular signaling to control processes such as hematopoiesis, immune function, and cellular growth and development. One key mechanism that regulates signaling from cytokine receptors is through receptor endocytosis. In this mini-review, we describe recent advances in endocytic regulations of cytokine receptors, focusing on new paradigms by which PI3K controls receptor endocytosis through both kinase activity-dependent and -independent mechanisms. These advances underscore the notion that the p85 regulatory subunit of PI3K has functions beyond regulating PI3K kinase activity, and that PI3K plays both positive and negative roles in receptor signaling. On the one hand, the PI3K/Akt pathway controls various aspects downstream of cytokine receptors. On the other hand, it stimulates receptor endocytosis and downregulation, thus contributing to signaling attenuation.
Collapse
Affiliation(s)
- Ping-hung Chen
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Huiyu Yao
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lily Jun-shen Huang
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- *Correspondence: Lily Jun-shen Huang,
| |
Collapse
|
28
|
Shi S, Liang D, Bao M, Xie Y, Xu W, Wang L, Wang Z, Qiao Z. Gx-50 Inhibits Neuroinflammation via α7 nAChR Activation of the JAK2/STAT3 and PI3K/AKT Pathways. J Alzheimers Dis 2016; 50:859-71. [PMID: 26836188 DOI: 10.3233/jad-150963] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Recent studies have revealed that the α7 nicotinic acetylcholine receptor (α7 nAChR) is a critical link between inflammation and neurodegeneration, which is closely associated with Alzheimer's disease (AD). The JAK2/STAT3 and PI3K/AKT signaling pathways contribute to the neuroprotective and anti-inflammatory effects of α7nAChR. Our previous studies have shown that treatment with gx-50 improves cognitive function and is neuroprotective. Here, we investigated the effect of gx-50 on α7 nAChR and Aβ-induced inflammation in microglia. First, the binding affinity of gx-50 to α7 nAChR was examined using the fluorescence-based Octet RED system, and the expression of α7 nAChR was detected using real-time PCR and western blotting. We also investigated downstream events of α7 nAChR activity, including the translocation of p-STAT3 and the phosphorylation of JAK2, STAT3, PI3K, and AKT. Finally, the effect of gx-50 on Aβ-induced inflammation via α7 nAChR-mediated signaling pathways was investigated using cytokine assays. The results showed that gx-50 is able to act as a specific ligand to activate α7 nAChR, which then upregulates the JAK2/STAT3 and PI3K/AKT signaling pathways to inhibit the secretions of pro-inflammatory cytokines, such as IL-1β. In conclusion, the results suggest that gx-50 could inhibit the Aβ-induced inflammatory response in microglia via α7 nAChR activity, which might be a successful therapeutic target against AD.
Collapse
|
29
|
Yu Z, Wan Y, Liu Y, Yang J, Li L, Zhang W. Curcumin induced apoptosis via PI3K/Akt-signalling pathways in SKOV3 cells. PHARMACEUTICAL BIOLOGY 2016; 54:2026-2032. [PMID: 26911246 DOI: 10.3109/13880209.2016.1139601] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Context Curcumin is widely used in China and India as a traditional herb but additional work is required to ascertain the folkloric claim of its antitumour and antioxidant activities. Objective The present study determines the antitumour effect of curcumin against SKOV3 cell growth. Materials and methods SKOV3 cells were incubated with curcumin (0, 20, 30 and 40 μM) for 72 h. The antiproliferative activity and the apoptosis rate were measured by MTT and flow cytometry. Expression of PI3K, T-Akt and p-Akt proteins was measured by western blotting. Results The administration of curcumin (0, 20, 30 and 40 μM) inhibits SKOV3 cell growth (IC50 value= 24.8 μM) and increased apoptosis (32.5 and 85.7%). The activity of SKOV3 cell invasion (98.2 and 19.4%) was also decreased by curcumin administration (p < 0.05). Results of western blot analysis confirmed that the expression of p-Akt protein was decreased by curcumin (p < 0.05). It was also found that a high dose of curcumin (40 μM) can cause stronger antitumour activity (80.4%). Conclusion Our results suggest that the curcumin induced SKOV3 apoptosis via modulation of the PI3K/Akt-signalling pathway.
Collapse
Affiliation(s)
- Zeshun Yu
- a Department of Oncology , Binzhou Medical University Hospital , Shandong , China
| | - Yingjun Wan
- b Department of Oncology , Binzhou PEOPLE'S Hospital , Shandong , China
| | - Yanni Liu
- c Department of Gynecology , Binzhou Medical University Hospital , Shandong , China
| | - Jing Yang
- a Department of Oncology , Binzhou Medical University Hospital , Shandong , China
| | - Lei Li
- a Department of Oncology , Binzhou Medical University Hospital , Shandong , China
| | - Weiming Zhang
- a Department of Oncology , Binzhou Medical University Hospital , Shandong , China
| |
Collapse
|
30
|
Zwang NA, Zhang R, Germana S, Fan MY, Hastings WD, Cao A, Turka LA. Selective Sparing of Human Tregs by Pharmacologic Inhibitors of the Phosphatidylinositol 3-Kinase and MEK Pathways. Am J Transplant 2016; 16:2624-38. [PMID: 27017850 PMCID: PMC5007157 DOI: 10.1111/ajt.13805] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 02/29/2016] [Accepted: 03/20/2016] [Indexed: 01/25/2023]
Abstract
Phosphatidylinositol 3-kinase (PI3K) and mitogen-activated protein kinase/extracellular signal-regulated (MEK) signaling are central to the survival and proliferation of many cell types. Multiple lines of investigation in murine models have shown that control of the PI3K pathway is particularly important for regulatory T cell (Treg) stability and function. PI3K and MEK inhibitors are being introduced into the clinic, and we hypothesized that pharmacologic inhibition of PI3K, and possibly MEK, in mixed cultures of human mononuclear cells would preferentially affect CD4(+) and CD8(+) lymphocytes compared with Tregs. We tested this hypothesis using four readouts: proliferation, activation, functional suppression, and signaling. Results showed that Tregs were less susceptible to inhibition by both δ and α isoform-specific PI3K inhibitors and by an MEK inhibitor compared with their conventional CD4(+) and CD8(+) counterparts. These studies suggest less functional reliance on PI3K and MEK signaling in Tregs compared with conventional CD4(+) and CD8(+) lymphocytes. Therefore, the PI3K and MEK pathways are attractive pharmacologic targets for transplantation and treatment of autoimmunity.
Collapse
Affiliation(s)
- N. A. Zwang
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA
- Massachusetts General Hospital/Brigham and Women’s Hospital Nephrology Joint Fellowship Program, Boston, MA
| | - R. Zhang
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA
| | - S. Germana
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA
| | - M. Y. Fan
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA
| | | | - A. Cao
- Novartis Pharmaceuticals, Cambridge, MA
| | - L. A. Turka
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
31
|
Silveira AB, Laranjeira ABA, Rodrigues GOL, Leal PC, Cardoso BA, Barata JT, Yunes RA, Zanchin NIT, Brandalise SR, Yunes JA. PI3K inhibition synergizes with glucocorticoids but antagonizes with methotrexate in T-cell acute lymphoblastic leukemia. Oncotarget 2016; 6:13105-18. [PMID: 25869207 PMCID: PMC4537002 DOI: 10.18632/oncotarget.3524] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 03/09/2015] [Indexed: 01/22/2023] Open
Abstract
The PI3K pathway is frequently hyperactivated in primary T-cell acute lymphoblastic leukemia (T-ALL) cells. Activation of the PI3K pathway has been suggested as one mechanism of glucocorticoid resistance in T-ALL, and patients harboring mutations in the PI3K negative regulator PTEN may be at increased risk of induction failure and relapse. By gene expression microarray analysis of T-ALL cells treated with the PI3K inhibitor AS605240, we identified Myc as a prominent downstream target of the PI3K pathway. A significant association was found between the AS605240 gene expression signature and that of glucocorticoid resistance and relapse in T-ALL. AS605240 showed anti-leukemic activity and strong synergism with glucocorticoids both in vitro and in a NOD/SCID xenograft model of T-ALL. In contrast, PI3K inhibition showed antagonism with methotrexate and daunorubicin, drugs that preferentially target dividing cells. This antagonistic interaction, however, could be circumvented by the use of correct drug scheduling schemes. Our data indicate the potential benefits and difficulties for the incorporation of PI3K inhibitors in T-ALL therapy.
Collapse
Affiliation(s)
| | | | | | - Paulo César Leal
- Departamento de Química, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Bruno António Cardoso
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - João Taborda Barata
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Rosendo Augusto Yunes
- Departamento de Química, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | | | | | - José Andrés Yunes
- Laboratório de Biologia Molecular, Centro Infantil Boldrini, Campinas, SP, Brazil.,Departamento de Genética Médica, Faculdade de Ciências Médicas, UNICAMP, Campinas, SP, Brazil
| |
Collapse
|
32
|
Cheung LW, Mills GB. Targeting therapeutic liabilities engendered by PIK3R1 mutations for cancer treatment. Pharmacogenomics 2016; 17:297-307. [PMID: 26807692 DOI: 10.2217/pgs.15.174] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The regulatory subunit of PI3K, p85α (encoded by PIK3R1), binds, stabilizes and inhibits the PI3K p110 catalytic subunit. Functional characterization of PIK3R1 mutations has identified not only hypomorphs with reduced inhibition of p110, but also hypomorphs and dominant negative mutants that disrupt a novel regulatory role of p85α on PTEN or neomorphs that activate unexpected signaling pathways. The diverse phenotypic spectrum of these PIK3R1 driver mutations underscores the need for different treatment strategies targeting tumors harboring these mutations. This article describes the functional consequences of the spectrum of PIK3R1 driver mutations and therapeutic liabilities they may engender.
Collapse
Affiliation(s)
- Lydia Wt Cheung
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gordon B Mills
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Khalifa Bin Zayed Al Nahyan Institute of Personalized Cancer Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
33
|
An R, Zhao L, Xi C, Li H, Shen G, Liu H, Zhang S, Sun L. Melatonin attenuates sepsis-induced cardiac dysfunction via a PI3K/Akt-dependent mechanism. Basic Res Cardiol 2015; 111:8. [PMID: 26671026 DOI: 10.1007/s00395-015-0526-1] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 11/24/2015] [Indexed: 12/21/2022]
Abstract
Myocardial dysfunction is an important manifestation of sepsis. Previous studies suggest that melatonin is protective against sepsis. In addition, activation of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway has been reported to be beneficial in sepsis. However, the role of PI3K/Akt signaling in the protective effect of melatonin against sepsis-induced myocardial dysfunction remains unclear. Here, LY294002, a PI3K inhibitor, was used to investigate the role of PI3K/Akt signaling in mediating the effects of melatonin on sepsis-induced myocardial injury. Cecal ligation and puncture (CLP) surgery was used to establish a rat model of sepsis. Melatonin was administrated to rats intraperitoneally (30 mg/kg). The survival rate, measures of myocardial injury and cardiac performance, serum lactate dehydrogenase level, inflammatory cytokine levels, oxidative stress level, and the extent of myocardial apoptosis were assessed. The results suggest that melatonin administration after CLP surgery improved survival rates and cardiac function, attenuated myocardial injury and apoptosis, and decreased the serum lactate dehydrogenase level. Melatonin decreased the production of the inflammatory cytokines TNF-α, IL-1β, and HMGB1, increased anti-oxidant enzyme activity, and decreased the expression of markers of oxidative damage. Levels of phosphorylated Akt (p-Akt), unphosphorylated Akt (Akt), Bcl-2, and Bax were measured by Western blot. Melatonin increased p-Akt levels, which suggests Akt pathway activation. Melatonin induced higher Bcl-2 expression and lower Bax expression, suggesting inhibition of apoptosis. All protective effects of melatonin were abolished by LY294002, the PI3K inhibitor. In conclusion, our results demonstrate that melatonin mitigates myocardial injury in sepsis via PI3K/Akt signaling activation.
Collapse
Affiliation(s)
- Rui An
- Department of Radiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Lei Zhao
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Cong Xi
- Department of Neurology, Baoji City People's Hospital, Baoji, 721000, China
| | - Haixun Li
- Department of Radiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Guohong Shen
- Integrated Branch, Armed Police Corps Hospital of Shanxi Province, Taiyuan, 030006, China
| | - Haixiao Liu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Shumiao Zhang
- Department of Physiology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Lijun Sun
- Department of Radiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
34
|
Yin D, Yang X, Li H, Fan H, Zhang X, Feng Y, Stuart C, Hu D, Caudle Y, Xie N, Liu Z, LeSage G. β-Arrestin 2 Promotes Hepatocyte Apoptosis by Inhibiting Akt Protein. J Biol Chem 2015; 291:605-12. [PMID: 26582201 DOI: 10.1074/jbc.m115.655829] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Indexed: 01/12/2023] Open
Abstract
Recent studies reveal that multifunctional protein β-arrestin 2 (Arrb2) modulates cell apoptosis. Survival and various aspects of liver injury were investigated in WT and Arrb2 KO mice after bile duct ligation (BDL). We found that deficiency of Arrb2 enhances survival and attenuates hepatic injury and fibrosis. Following BDL, Arrb2-deficient mice as compared with WT controls displayed a significant reduction of hepatocyte apoptosis as demonstrated by the TUNEL assay. Following BDL, the levels of phospho-Akt and phospho-glycogen synthase kinase 3β (GSK3β) in the livers were significantly increased in Arrb2 KO compared with WT mice, although p-p38 increased in WT but not in Arrb2-deficient mice. Inhibition of GSK3β following BDL decreases hepatic apoptosis and decreased p-p38 in WT mice but not in Arrb2 KO mice. Activation of Fas receptor with Jo2 reduces phospho-Akt and increases apoptosis in WT cells and WT mice but not in Arrb2-deficient cells and Arrb2-deficient mice. Consistent with direct interaction of Arrb2 with and regulating Akt phosphorylation, the expression of a full-length or N terminus but not the C terminus of Arrb2 reduces Akt phosphorylation and coimmunoprecipates with Akt. These results reveal that the protective effect of deficiency of Arrb2 is due to loss of negative regulation of Akt due to BDL and decreased downstream GSK3β and p38 MAPK signaling pathways.
Collapse
Affiliation(s)
- Deling Yin
- From the Department of Internal Medicine, College of Medicine, East Tennessee State University, Johnson City, Tennessee 37604 and
| | - Xiaohua Yang
- From the Department of Internal Medicine, College of Medicine, East Tennessee State University, Johnson City, Tennessee 37604 and
| | - Hui Li
- From the Department of Internal Medicine, College of Medicine, East Tennessee State University, Johnson City, Tennessee 37604 and
| | - Huimin Fan
- the Department of Cardiothoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Xiaoli Zhang
- From the Department of Internal Medicine, College of Medicine, East Tennessee State University, Johnson City, Tennessee 37604 and
| | - Yimin Feng
- From the Department of Internal Medicine, College of Medicine, East Tennessee State University, Johnson City, Tennessee 37604 and
| | - Charles Stuart
- From the Department of Internal Medicine, College of Medicine, East Tennessee State University, Johnson City, Tennessee 37604 and
| | - Dan Hu
- From the Department of Internal Medicine, College of Medicine, East Tennessee State University, Johnson City, Tennessee 37604 and
| | - Yi Caudle
- From the Department of Internal Medicine, College of Medicine, East Tennessee State University, Johnson City, Tennessee 37604 and
| | - Nanchang Xie
- From the Department of Internal Medicine, College of Medicine, East Tennessee State University, Johnson City, Tennessee 37604 and
| | - Zhongmin Liu
- the Department of Cardiothoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Gene LeSage
- From the Department of Internal Medicine, College of Medicine, East Tennessee State University, Johnson City, Tennessee 37604 and
| |
Collapse
|
35
|
Toubiana J, Rossi AL, Belaidouni N, Grimaldi D, Pene F, Chafey P, Comba B, Camoin L, Bismuth G, Claessens YE, Mira JP, Chiche JD. Src-family-tyrosine kinase Lyn is critical for TLR2-mediated NF-κB activation through the PI 3-kinase signaling pathway. Innate Immun 2015; 21:685-97. [DOI: 10.1177/1753425915586075] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 03/17/2015] [Indexed: 12/13/2022] Open
Abstract
TLR2 has a prominent role in host defense against a wide variety of pathogens. Stimulation of TLR2 triggers MyD88-dependent signaling to induce NF-κB translocation, and activates a Rac1-PI 3-kinase dependent pathway that leads to transactivation of NF-κB through phosphorylation of the P65 NF-κB subunit. This transactivation pathway involves tyrosine phosphorylations. The role of the tyrosine kinases in TLR signaling is controversial, with discrepancies between studies using only chemical inhibitors and knockout mice. Here, we show the involvement of the tyrosine-kinase Lyn in TLR2-dependent activation of NF-κB in human cellular models, by using complementary inhibition strategies. Stimulation of TLR2 induces the formation of an activation cluster involving TLR2, CD14, PI 3-kinase and Lyn, and leads to the activation of AKT. Lyn-dependent phosphorylation of the p110 catalytic subunit of PI 3-kinase is essential to the control of PI 3-kinase biological activity upstream of AKT and thereby to the transactivation of NF-κB. Thus, Lyn kinase activity is crucial in TLR2-mediated activation of the innate immune response in human mononuclear cells.
Collapse
Affiliation(s)
- Julie Toubiana
- Department of Infection, Immunity and Inflammation, Institut Cochin, Inserm, U1016, Paris, France
- Université Paris Descartes, Paris, France
- Department of Pediatrics, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Anne-Lise Rossi
- Department of Infection, Immunity and Inflammation, Institut Cochin, Inserm, U1016, Paris, France
- Université Paris Descartes, Paris, France
| | - Nadia Belaidouni
- Department of Infection, Immunity and Inflammation, Institut Cochin, Inserm, U1016, Paris, France
| | - David Grimaldi
- Department of Infection, Immunity and Inflammation, Institut Cochin, Inserm, U1016, Paris, France
- Université Paris Descartes, Paris, France
- Medical Intensive Care Unit, Hôpital Cochin, AP-HP, Paris, France
| | - Frederic Pene
- Department of Infection, Immunity and Inflammation, Institut Cochin, Inserm, U1016, Paris, France
- Université Paris Descartes, Paris, France
- Medical Intensive Care Unit, Hôpital Cochin, AP-HP, Paris, France
| | - Philippe Chafey
- Department of Infection, Immunity and Inflammation, Institut Cochin, Inserm, U1016, Paris, France
- Université Paris Descartes, Paris, France
| | - Béatrice Comba
- Department of Infection, Immunity and Inflammation, Institut Cochin, Inserm, U1016, Paris, France
| | - Luc Camoin
- Department of Infection, Immunity and Inflammation, Institut Cochin, Inserm, U1016, Paris, France
- Université Paris Descartes, Paris, France
| | - Georges Bismuth
- Department of Infection, Immunity and Inflammation, Institut Cochin, Inserm, U1016, Paris, France
- Université Paris Descartes, Paris, France
| | - Yann-Erick Claessens
- Department of Infection, Immunity and Inflammation, Institut Cochin, Inserm, U1016, Paris, France
- Université Paris Descartes, Paris, France
| | - Jean-Paul Mira
- Department of Infection, Immunity and Inflammation, Institut Cochin, Inserm, U1016, Paris, France
- Université Paris Descartes, Paris, France
- Medical Intensive Care Unit, Hôpital Cochin, AP-HP, Paris, France
| | - Jean-Daniel Chiche
- Department of Infection, Immunity and Inflammation, Institut Cochin, Inserm, U1016, Paris, France
- Université Paris Descartes, Paris, France
- Medical Intensive Care Unit, Hôpital Cochin, AP-HP, Paris, France
| |
Collapse
|
36
|
Mounayar M, Kefaloyianni E, Smith B, Solhjou Z, Maarouf OH, Azzi J, Chabtini L, Fiorina P, Kraus M, Briddell R, Fodor W, Herrlich A, Abdi R. PI3kα and STAT1 Interplay Regulates Human Mesenchymal Stem Cell Immune Polarization. Stem Cells 2015; 33:1892-901. [PMID: 25753288 PMCID: PMC4976699 DOI: 10.1002/stem.1986] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 01/17/2015] [Accepted: 02/10/2015] [Indexed: 12/14/2022]
Abstract
The immunomodulatory capacity of mesenchymal stem cells (MSCs) is critical for their use in therapeutic applications. MSC response to specific inflammatory cues allows them to switch between a proinflammatory (MSC1) or anti-inflammatory (MSC2) phenotype. Regulatory mechanisms controlling this switch remain to be defined. One characteristic feature of MSC2 is their ability to respond to IFNγ with induction of indoleamine 2,3-dioxygenase (IDO), representing the key immunoregulatory molecule released by human MSC. Here, we show that STAT1 and PI3Kα pathways interplay regulates IFNγ-induced IDO production in MSC. Chemical phosphoinositide 3-kinase (PI3K) pan-inhibition, PI3Kα-specific inhibition or shRNA knockdown diminished IFNγ-induced IDO production. This effect involved PI3Kα-mediated upregulation of STAT1 protein levels and phosphorylation at Ser727. Overexpression of STAT1 or of a constitutively active PI3Kα mutant failed to induce basal IDO production, but shifted MSC into an MSC2-like phenotype by strongly enhancing IDO production in response to IFNγ as compared to controls. STAT1 overexpression strongly enhanced MSC-mediated T-cell suppression. The same effect could be induced using short-term pretreatment of MSC with a chemical inhibitor of the counter player of PI3K, phosphatase and tensin homolog. Finally, downregulation of STAT1 abrogated the immunosuppressive capacity of MSC. Our results for the first time identify critical upstream signals for the induced production of IDO in MSCs that could be manipulated therapeutically to enhance their immunosuppressive phenotype.
Collapse
Affiliation(s)
- Marwan Mounayar
- Transplantation Research Center, Brigham and Women’s and Children’s Hospital, Boston, Massachusetts, USA
| | - Eirini Kefaloyianni
- Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Brian Smith
- Transplantation Research Center, Brigham and Women’s and Children’s Hospital, Boston, Massachusetts, USA
| | - Zhabiz Solhjou
- Transplantation Research Center, Brigham and Women’s and Children’s Hospital, Boston, Massachusetts, USA
| | - Omar H. Maarouf
- Transplantation Research Center, Brigham and Women’s and Children’s Hospital, Boston, Massachusetts, USA
| | - Jamil Azzi
- Transplantation Research Center, Brigham and Women’s and Children’s Hospital, Boston, Massachusetts, USA
| | - Lola Chabtini
- Transplantation Research Center, Brigham and Women’s and Children’s Hospital, Boston, Massachusetts, USA
| | - Paolo Fiorina
- Transplantation Research Center, Brigham and Women’s and Children’s Hospital, Boston, Massachusetts, USA
| | - Morey Kraus
- Viacord LLC, A PerkinElmer Company, Cambridge, Massachusetts, USA
| | - Robert Briddell
- ViaCord Processing Lab, ViaCord LLC, A PerkinElmer Company, Hebron, Kentucky, USA
| | | | - Andreas Herrlich
- Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Reza Abdi
- Transplantation Research Center, Brigham and Women’s and Children’s Hospital, Boston, Massachusetts, USA
| |
Collapse
|
37
|
Molecular mechanism underlying chemoprotective effects of Ganoderma atrum polysaccharide in cyclophosphamide-induced immunosuppressed mice. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.03.015] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
38
|
Beretta M, Bauer M, Hirsch E. PI3K signaling in the pathogenesis of obesity: The cause and the cure. Adv Biol Regul 2015; 58:1-15. [PMID: 25512233 DOI: 10.1016/j.jbior.2014.11.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 11/21/2014] [Accepted: 11/21/2014] [Indexed: 06/04/2023]
Abstract
With the steady rise in the incidence of obesity and its associated comorbidities, in the last decades research aimed at understanding molecular mechanisms that control body weight has gained new interest. Fat gain is frequently associated with chronic adipose tissue inflammation and with peripheral as well as central metabolic derangements, resulting in an impaired hypothalamic regulation of energy homeostasis. Recent attention has focused on the role of phosphatidylinositol 3-kinase (PI3K) in both immune and metabolic response pathways, being involved in the pathophysiology of obesity and its associated metabolic diseases. In this review, we focus on distinct PI3K isoforms, especially class I PI3Ks, mediating inflammatory cells recruitment to the enlarged fat as well as intracellular responses to key hormonal regulators of fat storage, both in adipocytes and in the central nervous system. This integrated view of PI3K functions may ultimately help to develop new therapeutic interventions for the treatment of obesity.
Collapse
Affiliation(s)
- Martina Beretta
- Molecular Biotechnology Center, University of Torino, Torino, Italy; Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
| | - Michael Bauer
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
| | - Emilio Hirsch
- Molecular Biotechnology Center, University of Torino, Torino, Italy.
| |
Collapse
|
39
|
Yu Q, Nie SP, Wang JQ, Huang DF, Li WJ, Xie MY. Toll-like receptor 4 mediates the antitumor host response induced by Ganoderma atrum polysaccharide. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:517-525. [PMID: 25549720 DOI: 10.1021/jf5041096] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
The aim of this study is to investigate the role of Toll-like receptor (TLR) 4 in Ganoderma atrum polysaccharide (PSG-1)-induced antitumor activity. In vitro, the apoptosis rate of S-180 cells was increased in PSG-1-induced peritoneal macrophage derived from C3H/HeN (wild-type) mice, but not from C3H/HeJ (TLR4-deficient) mice. In the S-180 tumor model, phagocytosis, NO and ROS release, phosphorylation of MAPKs and Akt, and expression of NF-κB were increased by PSG-1 in peritoneal macrophage derived from C3H/HeN mice. Furthermore, PSG-1 elevated Th1 cytokine production and enhanced the cytotoxic activity of CTL and NK cells in C3H/HeN mice. In addition, PSG-1 decreased the tumor weight and increased the apoptosis rate and caspase-3 and caspase-9 activities of tumor derived from the C3H/HeN mice. However, none of these activities were observed in C3H/HeJ mice. In summary, these findings demonstrated that the antitumor activity of PSG-1 is mediated by TLR4.
Collapse
Affiliation(s)
- Qiang Yu
- State Key Laboratory of Food Science and Technology, Nanchang University , 235 Nanjing East Road, Nanchang 330047, China
| | | | | | | | | | | |
Collapse
|
40
|
Ha TS, Hong EJ, Han GD. Diabetic conditions downregulate the expression of CD2AP in podocytes via PI3-K/Akt signalling. Diabetes Metab Res Rev 2015; 31:50-60. [PMID: 24846128 DOI: 10.1002/dmrr.2562] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 07/23/2012] [Accepted: 09/10/2012] [Indexed: 12/22/2022]
Abstract
BACKGROUND Proteinuria is typically accompanied by structural and compositional changes of the foot processes and of the slit diaphragms between podocytes. CD2-associated protein (CD2AP) in podocytes serves as an adaptor protein binding to nephrin and podocin, anchoring these slit diaphragm proteins to actin filaments of podocyte cytoskeleton and sending signals inward or outward. METHODS In the present study, we prepared streptozotocin-induced diabetic renal tissues and cultured podocytes in diabetic conditions to investigate podocyte phenotypical changes, including quantitative and distributional changes of CD2AP protein and search for the signalling mechanisms in diabetic conditions. We prepared cultured rat glomerular epithelial cells and mouse podocytes to study how high glucose and advanced glycosylation end products (AGE) induce phenotypical changes of cultured podocyte, under (1) normal glucose (5 mM, = control), (2) high glucose (30 mM), (3) AGE-added or (4) high glucose plus AGE-added conditions. RESULTS According to diabetic duration, density of CD2AP in renal tissue of experimental diabetic nephropathy became conglomerulated and diminished. In cultured podocytes, CD2AP co-localized with nephrin and zonula occludens-1 by confocal imaging. High glucose and high glucose plus AGE induced the relocalization and concentration of CD2AP at internal cytoplasmic and perinuclear areas of podocytes. High glucose plus AGE-added condition also decreased CD2AP protein amount and its mRNA expression compared with normal glucose or osmotic control conditions. In addition, LY294002, a phosphoinositide 3-kinase inhibitor, prevented the quantitative and distributional changes of CD2AP induced by high glucose and AGE. CONCLUSIONS These findings suggest that diabetic conditions induce the phenotypical changes of podocyte CD2AP possibly via phosphoinositide 3-kinase/Akt signalling.
Collapse
Affiliation(s)
- Tae-Sun Ha
- Department of Pediatrics, College of Medicine, Chungbuk National University, Cheongju, Korea
| | | | | |
Collapse
|
41
|
Roy R, Chauhan LKS, Das M, Tripathi A, Dwivedi PD. Phagocytic cells internalize ZnO particles by FcγII/III-receptor pathway. Immunobiology 2014; 219:746-55. [PMID: 25082708 DOI: 10.1016/j.imbio.2014.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Accepted: 06/03/2014] [Indexed: 10/25/2022]
Abstract
The present study investigates the process of internalization for bulk ZnO particles in macrophages, and further elucidates the underlying mechanism. Since macrophages are active phagocytes and phagocytosis is a size dependent phenomenon, therefore we hypothesized that bulk ZnO may internalize into macrophages by phagocytic pathways. Interestingly, the phagocytic activity got enhanced in bulk ZnO treated macrophages. Moreover, the bulk ZnO treated macrophages internalized via FcγR-II/III, complement and scavenger-receptor pathways. To confirm the specificity of phagocytic pathway, the uptake was also analyzed in splenocytes where phagocytic (monocytes) and non-phagocytic cells (lymphocytes) are present. It was observed that no significant uptake of bulk ZnO in case of lymphocytes whereas significant uptake in monocytes. Henceforth, our quest for uptake mechanisms also revealed that severe plasma membrane extensions (pseudopodia), FcγR clustering over the surface of macrophages and activation of FcγR signaling were the key players for bulk ZnO uptake; whereas clathrin or caveolae mediated endocytic pathways contributed less. Uptake of these particles was further strengthened by the ZnO-induced activation of the Src-kinase p-Lyn, phospho-tyrosine kinases Syk (spleen tyrosine kinase), p-PLC-γ and PI3K (phosphatidylinositol 3-kinase). Our findings illustrate that the phagocytic nature of macrophages could have led to higher uptake of bulk ZnO.
Collapse
Affiliation(s)
- Ruchi Roy
- Food, Drug and Chemical Toxicology Group, Indian Institute of Toxicology Research, M.G. Marg. Post Box No. 80, Lucknow 226001, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - L K S Chauhan
- Electron Microscopy Facility, Indian Institute of Toxicology Research, M.G. Marg. Post Box No. 80, Lucknow 226001, India
| | - Mukul Das
- Food, Drug and Chemical Toxicology Group, Indian Institute of Toxicology Research, M.G. Marg. Post Box No. 80, Lucknow 226001, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Anurag Tripathi
- Food, Drug and Chemical Toxicology Group, Indian Institute of Toxicology Research, M.G. Marg. Post Box No. 80, Lucknow 226001, India.
| | - Premendra D Dwivedi
- Food, Drug and Chemical Toxicology Group, Indian Institute of Toxicology Research, M.G. Marg. Post Box No. 80, Lucknow 226001, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi, India.
| |
Collapse
|
42
|
Perez DA, Vago JP, Athayde RM, Reis AC, Teixeira MM, Sousa LP, Pinho V. Switching off key signaling survival molecules to switch on the resolution of inflammation. Mediators Inflamm 2014; 2014:829851. [PMID: 25136148 PMCID: PMC4127222 DOI: 10.1155/2014/829851] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 06/02/2014] [Accepted: 07/01/2014] [Indexed: 01/07/2023] Open
Abstract
Inflammation is a physiological response of the immune system to injury or infection but may become chronic. In general, inflammation is self-limiting and resolves by activating a termination program named resolution of inflammation. It has been argued that unresolved inflammation may be the basis of a variety of chronic inflammatory diseases. Resolution of inflammation is an active process that is fine-tuned by the production of proresolving mediators and the shutdown of intracellular signaling molecules associated with cytokine production and leukocyte survival. Apoptosis of leukocytes (especially granulocytes) is a key element in the resolution of inflammation and several signaling molecules are thought to be involved in this process. Here, we explore key signaling molecules and some mediators that are crucial regulators of leukocyte survival in vivo and that may be targeted for therapeutic purposes in the context of chronic inflammatory diseases.
Collapse
Affiliation(s)
- Denise Alves Perez
- Laboratório de Resolução da Resposta Inflamatória, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil
| | - Juliana Priscila Vago
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil
- Laboratório de Sinalização inflamação, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil
| | - Rayssa Maciel Athayde
- Laboratório de Resolução da Resposta Inflamatória, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil
| | - Alesandra Corte Reis
- Laboratório de Resolução da Resposta Inflamatória, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil
| | - Mauro Martins Teixeira
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil
| | - Lirlândia Pires Sousa
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil
- Laboratório de Sinalização inflamação, Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil
| | - Vanessa Pinho
- Laboratório de Resolução da Resposta Inflamatória, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil
| |
Collapse
|
43
|
ZSTK474, a novel PI3K inhibitor, modulates human CD14+ monocyte-derived dendritic cell functions and suppresses experimental autoimmune encephalomyelitis. J Mol Med (Berl) 2014; 92:1057-68. [PMID: 24844601 DOI: 10.1007/s00109-014-1158-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 04/16/2014] [Accepted: 04/17/2014] [Indexed: 10/25/2022]
Abstract
ZSTK474 [2-(2-difluoromethylbenzimidazol-1-yl)-4,6-dimorpholino-1,3,5-triazine] is a novel phosphatidylinositol 3-kinase (PI3K) inhibitor that exhibits potent antitumor effects. Recent studies have shown that ZSTK474 is also with anti-inflammatory properties in collagen-induced arthritis. However, the effects of ZSTK474 on dendritic cells and inflammatory Th17 cell-mediated autoimmune central nervous system inflammation are not understood. In this study, we demonstrated that ZSTK474 suppressed human CD14(+) monocyte-derived dendritic cell differentiation, maturation, and endocytosis, and further inhibited the stimulatory function of mature dendritic cell on allogeneic T cell proliferation. In addition, ZSTK474 inhibited the expression of dendritic cell-derived Th1 and Th17 cells polarizing cytokines interferon-γ/interleukin (IL)-12 and IL-6/IL-23. Furthermore, our results indicated that the in vivo administration of ZSTK474, which targets the dendritic cell and inflammatory Th1 and Th17 cell, led to a reduction of clinical score, central nervous system inflammation, and demyelination of mouse experimental autoimmune encephalomyelitis. Therefore, ZSTK474 significantly suppressed the human CD14(+) monocyte-derived dendritic cell functions and ameliorated mouse experimental autoimmune encephalomyelitis. We further found that ZSTK474 inhibited the phosphorylation of PI3K downstream signaling Akt and glycogen synthase kinase 3 beta in the dendritic cell. These data suggested that ZSTK474 exerted potent anti-inflammatory and immunosuppressive properties via PI3K signaling and may serve as a potential therapeutic drug for multiple sclerosis and other autoimmune inflammatory diseases. Key messages: STK474 inhibits dendritic cell (DC) differentiation and maturation. ZSTK474 inhibits DC-derived Th1 and Th17-polarizing cytokines. ZSTK474 ameliorates EAE and suppresses DCs, Th1, and Th17 cells in EAE. ZSTK474 reduces CNS inflammation and demyelination of EAE mice. ZSTK474 could be a potential therapeutic drug for multiple sclerosis.
Collapse
|
44
|
Boyle DL, Kim HR, Topolewski K, Bartok B, Firestein GS. Novel phosphoinositide 3-kinase δ,γ inhibitor: potent anti-inflammatory effects and joint protection in models of rheumatoid arthritis. J Pharmacol Exp Ther 2014; 348:271-80. [PMID: 24244039 DOI: 10.1124/jpet.113.205955] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Phosphoinositide 3-kinases γ and δ (PI3Kγ and PI3Kδ) are expressed in rheumatoid arthritis (RA) synovium and regulate innate and adaptive immune responses. We determined the effect of a potent PI3Kδ,γ inhibitor, IPI-145, in two preclinical models of RA. IPI-145 was administered orally in rat adjuvant-induced arthritis (AA) and intraperitoneally in mouse collagen-induced arthritis (CIA). Efficacy was assessed by paw swelling, clinical scores, histopathology and radiography, and microcomputed tomography scanning. Gene expression and Akt phosphorylation in joint tissues were determined by quantitative real-time polymerase chain reaction and Western blot analysis. Serum concentrations of anti-type II collagen (CII) IgG and IgE were measured by immunoassay. T-cell responses to CII were assayed using thymidine incorporation and immunoassay. IPI-145 significantly reduced arthritis severity in both RA models using dosing regimens initiated before onset of clinical disease. Treatment of established arthritis with IPI-145 in AA, but not CIA, significantly decreased arthritis progression. In AA, histology scores, radiographic joint damage, and matrix metalloproteinase (MMP)-13 expression were reduced in IPI-145-treated rats. In CIA, joint histology scores and expression of MMP-3 and MMP-13 mRNA were lower in the IPI-145 early treatment group than in the vehicle group. The ratio of anti-CII IgG2a to total IgG in CIA was modestly reduced. Interleukin-17 production in response to CII was decreased in the IPI-145-treated group, suggesting an inhibitory effect on T-helper cell 17 differentiation. These data show that PI3Kδ,γ inhibition suppresses inflammatory arthritis, as well as bone and cartilage damage, through effects on innate and adaptive immunity and that IPI-145 is a potential therapy for RA.
Collapse
MESH Headings
- Administration, Oral
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/administration & dosage
- Anti-Inflammatory Agents, Non-Steroidal/therapeutic use
- Antirheumatic Agents/administration & dosage
- Antirheumatic Agents/therapeutic use
- Arthritis, Rheumatoid/drug therapy
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/pathology
- Cells, Cultured
- Disease Models, Animal
- Drugs, Investigational/administration & dosage
- Drugs, Investigational/therapeutic use
- Enzyme Inhibitors/administration & dosage
- Enzyme Inhibitors/therapeutic use
- Injections, Intraperitoneal
- Interleukin-17/antagonists & inhibitors
- Interleukin-17/genetics
- Interleukin-17/metabolism
- Isoenzymes/antagonists & inhibitors
- Isoenzymes/metabolism
- Isoquinolines/administration & dosage
- Isoquinolines/therapeutic use
- Joints/drug effects
- Joints/immunology
- Joints/metabolism
- Joints/pathology
- Male
- Mice
- Mice, Inbred DBA
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphoinositide-3 Kinase Inhibitors
- Purines/administration & dosage
- Purines/therapeutic use
- Random Allocation
- Rats
- Rats, Inbred Lew
- Synovial Membrane/drug effects
- Synovial Membrane/enzymology
- Synovial Membrane/immunology
- Synovial Membrane/pathology
- Th17 Cells/drug effects
- Th17 Cells/immunology
- Th17 Cells/metabolism
Collapse
Affiliation(s)
- David L Boyle
- Division of Rheumatology, Allergy, and Immunology, School of Medicine, University of California San Diego, La Jolla, California
| | | | | | | | | |
Collapse
|
45
|
Nobiletin, a polymethoxy flavonoid, exerts anti-allergic effect by suppressing activation of phosphoinositide 3-kinase. J Funct Foods 2014. [DOI: 10.1016/j.jff.2013.12.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
|
46
|
Tetramethylpyrazine analogue CXC195 protects against cerebral ischemia/reperfusion-induced apoptosis through PI3K/Akt/GSK3β pathway in rats. Neurochem Int 2014; 66:27-32. [DOI: 10.1016/j.neuint.2014.01.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 12/24/2013] [Accepted: 01/09/2014] [Indexed: 12/26/2022]
|
47
|
Morohaku K, Hoshino Y, Sasada H, Sato E. Incorporation of phosphatase inhibitor in culture prompts growth initiation of isolated non-growing oocytes. PLoS One 2013; 8:e77533. [PMID: 24223714 PMCID: PMC3817191 DOI: 10.1371/journal.pone.0077533] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2013] [Accepted: 09/03/2013] [Indexed: 12/31/2022] Open
Abstract
In vitro folliculogenesis of primordial and early preantral follicles is necessary for increment of reproductive efficiency in domestic animals, humans and endangered species. Recent study in phosphatase and tensin homolog (Pten) -knockout mice has revealed that this phosphatase acts as an inhibitory factor in follicle activation of primordial pool with the resultant inhibition of oocyte growth. To test in vitro effect of a phosphatase inhibitor on growth initiation of isolated non-growing oocytes in neonatal ovaries, we applied a specific inhibitor (bpV (HOpic)) for PTEN in culturing system. Non-growing oocytes isolated from the ovaries of newborn BDF1 (C57BL/6 × DBA/2) pups were divided to four culture groups. Five days after culture, the oocytes in 14 μmol/l bpV only, 14 μmol/l bpV plus 100 ng/ml Kit Ligand (KL), and 100 ng/ml KL groups showed significantly (P<0.05) growth (19.3±0.55, 25.8±0.53 and 21.6±0.29 μm, respectively) compared with that of the control (no additive) (16.9±0.53 μm). In addition, western blotting in those groups showed enhanced expression of phosphorylated Akt. In conclusion, we clearly demonstrate that isolated non-growing oocytes develop in phosphatase inhibitor, especially to PTEN, incorporated culturing system, and show first as we know that oocytes with zona Pellucidae can be obtained in vitro from isolated non-growing oocytes.
Collapse
Affiliation(s)
- Kanako Morohaku
- Laboratory of Animal Reproduction, Graduate School of Agriculture Science, Tohoku University, Sendai, Japan
- Laboratory of Animal Reproduction, School of Veterinary Medicine, Kitasato University, Towada, Japan
- * E-mail:
| | - Yumi Hoshino
- Laboratory of Animal Reproduction, Graduate School of Agriculture Science, Tohoku University, Sendai, Japan
| | - Hiroshi Sasada
- Laboratory of Animal Reproduction, Graduate School of Agriculture Science, Tohoku University, Sendai, Japan
- Laboratory of Animal Reproduction, School of Veterinary Medicine, Kitasato University, Towada, Japan
| | - Eimei Sato
- Laboratory of Animal Reproduction, Graduate School of Agriculture Science, Tohoku University, Sendai, Japan
| |
Collapse
|
48
|
Markopoulou A, Kyttaris VC. Small molecules in the treatment of systemic lupus erythematosus. Clin Immunol 2013; 148:359-68. [PMID: 23158694 PMCID: PMC3587286 DOI: 10.1016/j.clim.2012.09.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 09/18/2012] [Accepted: 09/22/2012] [Indexed: 12/17/2022]
Abstract
Advances in the understanding of the cellular biological events that underlie systemic lupus erythematosus (SLE) have led to the identification of key molecules and signaling pathways that are aberrantly expressed. The parallel development of small molecule drugs that inhibit or interfere with the specific perturbations identified, offers perspective for more rational, effective and less toxic therapy. In this review, we present data from preclinical and clinical studies of such emerging novel therapies with a particular focus on kinase inhibitors and other compounds that modulate signal transduction. Moreover, we highlight the use of chromatin-modifying medications, bringing attention to the central role of epigenetics in SLE pathogenesis.
Collapse
Affiliation(s)
- Anastasia Markopoulou
- Division of Rheumatology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
49
|
Shanware NP, Bray K, Abraham RT. The PI3K, metabolic, and autophagy networks: interactive partners in cellular health and disease. Annu Rev Pharmacol Toxicol 2013; 53:89-106. [PMID: 23294306 DOI: 10.1146/annurev-pharmtox-010611-134717] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
A fundamental imperative for mammalian cells is to coordinate cell metabolism and growth with environmentally induced stress. This review focuses on three highly integrated networks-the phosphoinositide 3-kinase (PI3K) signaling cascade, intermediate metabolism, and autophagy-that work together to maintain cellular homeostasis under basal conditions and to drive cell-mass accumulation and cell cycle progression in the presence of appropriate mitogenic stimuli. Dysfunction within any one of these networks results in compensatory responses from the other networks. These responses underpin several pathologies associated with major human diseases such as cancer. We discuss the PI3K, metabolism, and autophagy networks and provide selected insights into internetwork cross-talk mechanisms. In recognition of the extensive interactions observed in both healthy and diseased cells, we propose that the three networks be merged into a "metabolism-signaling supernetwork." A detailed understanding of this supernetwork will facilitate the development of novel therapies for cancer and other complex diseases.
Collapse
Affiliation(s)
- Naval P Shanware
- Oncology Research Unit, Pfizer Worldwide Research and Development, Pearl River, New York 10965, USA
| | | | | |
Collapse
|
50
|
McCurdy CE, Klemm DJ. Adipose tissue insulin sensitivity and macrophage recruitment: Does PI3K pick the pathway? Adipocyte 2013; 2:135-42. [PMID: 23991359 PMCID: PMC3756101 DOI: 10.4161/adip.24645] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 04/09/2013] [Accepted: 04/10/2013] [Indexed: 02/06/2023] Open
Abstract
In the United States, obesity is a burgeoning health crisis, with over 30% of adults and nearly 20% of children classified as obese. Insulin resistance, a common metabolic complication associated with obesity, significantly increases the risk of developing metabolic diseases such as hypertension, coronary heart disease, stroke, type 2 diabetes, and certain cancers. With the seminal finding that obese adipose tissue harbors cytokine secreting immune cells, obesity-related research over the past decade has focused on understanding adipocyte–macrophage crosstalk and its impact on systemic insulin sensitivity. Indeed, adipose tissue has emerged as a central mediator of obesity- and diet-induced insulin resistance. In this mini-review, we focus on a potential role of adipose tissue phosphoinositide 3-kinase (PI3K) as a point of convergence of cellular signaling pathways that integrates nutrient sensing and inflammatory signaling to regulate tissue insulin sensitivity.
Collapse
|