1
|
Seyrek K, Espe J, Reiss E, Lavrik IN. The Crosstalk of Apoptotic and Non-Apoptotic Signaling in CD95 System. Cells 2024; 13:1814. [PMID: 39513921 PMCID: PMC11545656 DOI: 10.3390/cells13211814] [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: 09/26/2024] [Revised: 10/24/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
The mechanisms of CD95 (Fas/APO-1)-mediated extrinsic apoptotic pathway in cancer cells have been extensively studied. The majority of human cells express CD95, but not all these cells can induce extrinsic apoptosis. Accumulating evidence has shown that CD95 is a multifunctional protein, and its stimulation can also elicit non-apoptotic or even survival signals. It has become clear that under certain cellular contexts, due to the various checkpoints, CD95 activation can trigger both apoptotic and non-apoptotic signals. The crosstalk of death and survival signals may occur at different levels of signal transduction. The strength of the CD95 stimulation, initial levels of anti-apoptotic proteins, and posttranslational modifications of the core DISC components have been proposed to be the most important factors in the life/death decisions at CD95. Successful therapeutic targeting of CD95 signaling pathways will require a better understanding of the crosstalk between CD95-induced apoptotic and cell survival pathways. In this review, in order to gain a systematic understanding of the crosstalk between CD95-mediated apoptosis and non-apoptotic signaling, we will discuss these issues in a step-by-step way.
Collapse
Affiliation(s)
| | | | | | - Inna N. Lavrik
- Translational Inflammation Research, Medical Faculty, Otto von Guericke University Magdeburg, 39106 Magdeburg, Germany; (K.S.); (J.E.); (E.R.)
| |
Collapse
|
2
|
Biggi AFB, Silvestre RN, Tirapelle MC, de Azevedo JTC, García HDM, Henrique Dos Santos M, de Lima SCG, de Souza LEB, Covas DT, Malmegrim KCR, Figueiredo ML, Picanço-Castro V. IL-27-engineered CAR.19-NK-92 cells exhibit enhanced therapeutic efficacy. Cytotherapy 2024; 26:1320-1330. [PMID: 38970613 DOI: 10.1016/j.jcyt.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 05/08/2024] [Accepted: 06/01/2024] [Indexed: 07/08/2024]
Abstract
Chimeric antigen receptor (CAR) engineering of natural killer (NK) cells has shown promising results in early-phase clinical studies. However, advancing CAR-NK cell therapeutic efficacy is imperative. In this study, we investigated the impact of a fourth-generation CD19-targeted CAR (CAR.19) coexpressing IL-27 on NK-92 cells. We observed a significant improvement in NK-92 cell proliferation and cytotoxicity activity against B-cell cancer cell lines, both in vitro and in a xenograft mouse B-cell lymphoma model. Our systematic transcriptome analysis of the activated NK-92 CAR variants further supports the potential of IL-27 in fourth-generation CARs to overcome limitations of NK cell-based targeted tumor therapies by providing essential growth and activation signals. Integrating IL-27 into CAR-NK cells emerges as a promising strategy to enhance their therapeutic potential and elicit robust responses against cancer cells. These findings contribute substantially to the mounting evidence supporting the potential of fourth-generation CAR engineering in advancing NK cell-based immunotherapies.
Collapse
Affiliation(s)
- Alison Felipe Bordini Biggi
- Center for Cell-based Therapy CTC, Regional Blood Center of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Renata Nacasaki Silvestre
- Center for Cell-based Therapy CTC, Regional Blood Center of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Mariane Cariati Tirapelle
- Center for Cell-based Therapy CTC, Regional Blood Center of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Julia Teixeira Cottas de Azevedo
- Center for Cell-based Therapy CTC, Regional Blood Center of Ribeirão Preto, University of São Paulo, São Paulo, Brazil; Department of Hemotherapy and Cellular Therapy, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | | | - Matheus Henrique Dos Santos
- Center for Cell-based Therapy CTC, Regional Blood Center of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Sarah Caroline Gomes de Lima
- Center for Cell-based Therapy CTC, Regional Blood Center of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | | | - Dimas Tadeu Covas
- Center for Cell-based Therapy CTC, Regional Blood Center of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Kelen Cristina Ribeiro Malmegrim
- Center for Cell-based Therapy CTC, Regional Blood Center of Ribeirão Preto, University of São Paulo, São Paulo, Brazil; Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Marxa L Figueiredo
- Department of Basic Medical Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Virginia Picanço-Castro
- Center for Cell-based Therapy CTC, Regional Blood Center of Ribeirão Preto, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
3
|
Qi Y, Wang Y, Yuan J, Xu Y, Pan H. Unveiling the therapeutic promise: exploring Lysophosphatidic Acid (LPA) signaling in malignant bone tumors for novel cancer treatments. Lipids Health Dis 2024; 23:204. [PMID: 38943207 PMCID: PMC11212261 DOI: 10.1186/s12944-024-02196-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/19/2024] [Indexed: 07/01/2024] Open
Abstract
Malignant bone tumors, including primary bone cancer and metastatic bone tumors, are a significant clinical challenge due to their high frequency of presentation, poor prognosis and lack of effective treatments and therapies. Bone tumors are often accompanied by skeletal complications such as bone destruction and cancer-induced bone pain. However, the mechanisms involved in bone cancer progression, bone metastasis and skeletal complications remain unclear. Lysophosphatidic acid (LPA), an intercellular lipid signaling molecule that exerts a wide range of biological effects mainly through specifically binding to LPA receptors (LPARs), has been found to be present at high levels in the ascites of bone tumor patients. Numerous studies have suggested that LPA plays a role in primary malignant bone tumors, bone metastasis, and skeletal complications. In this review, we summarize the role of LPA signaling in primary bone cancer, bone metastasis and skeletal complications. Modulating LPA signaling may represent a novel avenue for future therapeutic treatments for bone cancer, potentially improving patient prognosis and quality of life.
Collapse
Affiliation(s)
- Yichen Qi
- Huankui Academy, Nanchang University, Nanchang, 330031, China
- Neurological Institute of Jiangxi Province, Department of Neurology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, China
- Department of Neurology, Xiangya Hospital, Central South University, Jiangxi Hospital, National Regional Center for Neurological Diseases, No. 266 Fenghe North Avenue, Honggutan District, Nanchang, Jiangxi, 330038, P. R. China
| | - Yukai Wang
- School of Life Sciences, Nanchang University, Nanchang, 330031, China
- Neurological Institute of Jiangxi Province, Department of Neurology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, China
- Department of Neurology, Xiangya Hospital, Central South University, Jiangxi Hospital, National Regional Center for Neurological Diseases, No. 266 Fenghe North Avenue, Honggutan District, Nanchang, Jiangxi, 330038, P. R. China
| | - Jinping Yuan
- The First Clinical Medical College, Nanchang University, Nanchang, 330031, China
- Neurological Institute of Jiangxi Province, Department of Neurology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, China
- Department of Neurology, Xiangya Hospital, Central South University, Jiangxi Hospital, National Regional Center for Neurological Diseases, No. 266 Fenghe North Avenue, Honggutan District, Nanchang, Jiangxi, 330038, P. R. China
| | - Yufei Xu
- The First Clinical Medical College, Nanchang University, Nanchang, 330031, China
- Neurological Institute of Jiangxi Province, Department of Neurology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, China
- Department of Neurology, Xiangya Hospital, Central South University, Jiangxi Hospital, National Regional Center for Neurological Diseases, No. 266 Fenghe North Avenue, Honggutan District, Nanchang, Jiangxi, 330038, P. R. China
| | - Haili Pan
- Neurological Institute of Jiangxi Province, Department of Neurology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, China.
- Department of Neurology, Xiangya Hospital, Central South University, Jiangxi Hospital, National Regional Center for Neurological Diseases, No. 266 Fenghe North Avenue, Honggutan District, Nanchang, Jiangxi, 330038, P. R. China.
| |
Collapse
|
4
|
Wilson JJ, Wei J, Daamen AR, Sears JD, Bechtel E, Mayberry CL, Stafford GA, Bechtold L, Grammer AC, Lipsky PE, Roopenian DC, Chang CH. Glucose oxidation-dependent survival of activated B cells provides a putative novel therapeutic target for lupus treatment. iScience 2023; 26:107487. [PMID: 37636066 PMCID: PMC10448027 DOI: 10.1016/j.isci.2023.107487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/27/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
Aberrant metabolic demand is observed in immune/inflammatory disorders, yet the role in pathogenesis remains unclear. Here, we discover that in lupus, activated B cells, including germinal center B (GCB) cells, have remarkably high glycolytic requirement for survival over T cell populations, as demonstrated by increased metabolic activity in lupus-activated B cells compared to immunization-induced cells. The augmented reliance on glucose oxidation makes GCB cells vulnerable to mitochondrial ROS-induced oxidative stress and apoptosis. Short-term glycolysis inhibition selectively reduces pathogenic activated B in lupus-prone mice, extending their lifespan, without affecting T follicular helper cells. Particularly, BCMA-expressing GCB cells rely heavily on glucose oxidation. Depleting BCMA-expressing activated B cells with APRIL-based CAR-T cells significantly prolongs the lifespan of mice with severe autoimmune disease. These results reveal that glycolysis-dependent activated B and GCB cells, especially those expressing BCMA, are potentially key lupus mediators, and could be targeted to improve disease outcomes.
Collapse
Affiliation(s)
- John J. Wilson
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA
| | - Jian Wei
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Andrea R. Daamen
- AMPEL BioSolutions and the RILITE Research Institute, Charlottesville, VA 22902, USA
| | - John D. Sears
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Elaine Bechtel
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA
| | | | | | | | - Amrie C. Grammer
- AMPEL BioSolutions and the RILITE Research Institute, Charlottesville, VA 22902, USA
| | - Peter E. Lipsky
- AMPEL BioSolutions and the RILITE Research Institute, Charlottesville, VA 22902, USA
| | | | - Chih-Hao Chang
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469, USA
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA
| |
Collapse
|
5
|
Saha C, Li J, Sun X, Liu X, Huang G. A novel role of Fas in delaying cellular senescence. Heliyon 2023; 9:e13451. [PMID: 36825177 PMCID: PMC9941949 DOI: 10.1016/j.heliyon.2023.e13451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 01/26/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Fas-mediated apoptosis is a major player of many physiological and pathological cellular processes. Fas-regulated immune regulation exhibits either the beneficial or the harmful effects which is associated with the onset or development of immune disorders. Alterations in apoptosis may contribute to age-associated changes. However, the role of apoptosis in the ageing process remains ambiguous. Here we demonstrated Fas signaling-mediated premature senescence in young mouse embryonic fibroblast (MEF) cells. Activated Fas signaling by agonist Jo-2 resulted in declined senescence in young and aged MEFs. Premature senescence induced the early activation of senescence markers, including the increase in the percentage of SA-β-galactosidase (SA-β-gal) cells, the induction of p53 phosphorylation, and the enhanced expression of p16 and p21 protein and elevated IL-6 pro-inflammatory cytokine in the absence of Fas. The elevated production of reactive oxygen species (ROS) in Fas-deficient MEFs was associated with dysfunctional mitochondria. Further, we determined that the known ROS scavenger NAC (N-acetyl-l-cysteine) could reverse the process of premature senescence in absence of Fas. Therefore, this study signifies a novel role of Fas in the control of cellular senescence.
Collapse
Affiliation(s)
- Chaitrali Saha
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523808, China
| | - Jingyu Li
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523808, China,School of Medical Technology, Guangdong Medical University, Dongguan, 523808, China
| | - Xuerong Sun
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523808, China
| | - Xinguang Liu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523808, China
| | - Gonghua Huang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523808, China,Corresponding author.
| |
Collapse
|
6
|
Quilty F, Freeley M, Gargan S, Gilmer J, Long A. Deoxycholic acid induces proinflammatory cytokine production by model oesophageal cells via lipid rafts. J Steroid Biochem Mol Biol 2021; 214:105987. [PMID: 34438042 DOI: 10.1016/j.jsbmb.2021.105987] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/18/2021] [Accepted: 08/21/2021] [Indexed: 11/28/2022]
Abstract
The bile acid component of gastric refluxate has been implicated in inflammation of the oesophagus including conditions such as gastro-oesophageal reflux disease (GORD) and Barrett's Oesophagus (BO). Here we demonstrate that the hydrophobic bile acid, deoxycholic acid (DCA), stimulated the production of IL-6 and IL-8 mRNA and protein in Het-1A, a model of normal oesophageal cells. DCA-induced production of IL-6 and IL-8 was attenuated by pharmacologic inhibition of the Protein Kinase C (PKC), MAP kinase, tyrosine kinase pathways, by the cholesterol sequestering agent, methyl-beta-cyclodextrin (MCD) and by the hydrophilic bile acid, ursodeoxycholic acid (UDCA). The cholesterol-interacting agent, nystatin, which binds cholesterol without removing it from the membrane, synergized with DCA to induce IL-6 and IL-8. This was inhibited by the tyrosine kinase inhibitor genistein. DCA stimulated the phosphorylation of lipid raft component Src tyrosine kinase (Src). while knockdown of caveolin-1 expression using siRNA resulted in a decreased level of IL-8 production in response to DCA. Taken together, these results demonstrate that DCA stimulates IL-6 and IL-8 production in oesophageal cells via lipid raft-associated signaling. Inhibition of this process using cyclodextrins represents a novel therapeutic approach to the treatment of inflammatory diseases of the oesophagus including GORD and BO.
Collapse
Affiliation(s)
- Francis Quilty
- School of Pharmacy and Pharmaceutical Science, Trinity College Dublin, Dublin 2, Ireland; Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Michael Freeley
- School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Siobhan Gargan
- Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - John Gilmer
- School of Pharmacy and Pharmaceutical Science, Trinity College Dublin, Dublin 2, Ireland; Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Aideen Long
- Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin 8, Ireland.
| |
Collapse
|
7
|
Loss of GATA4 C-Terminus by p.S335X Mutation Modulates Coronary Artery Vascular Smooth Muscle Cell Phenotype. Mediators Inflamm 2021; 2021:3698386. [PMID: 34545275 PMCID: PMC8449727 DOI: 10.1155/2021/3698386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 08/17/2021] [Indexed: 11/29/2022] Open
Abstract
Coronary artery disease (CAD) has been the leading cause of morbidity and mortality worldwide, and its pathogenesis is closely related with the proliferation and migration of vascular smooth muscle cell (VSMC). We previously reported a truncated GATA4 protein lacking C-terminus induced by p.S335X mutation in cardiomyocyte from ventricular septal defect (VSD) patients. However, it is still unclear whether GATA4 p.S335X mutation could influence the development of CAD. GATA4 wild-type (WT) and p.S335X mutant (MU) overexpression plasmids were constructed and transfected transiently into rat coronary artery smooth muscle cell (RCSMC) to observe the proliferative and migratory abilities by MTS and wound healing assay, respectively. PCR array was used to preliminarily detect the expression of phenotypic modulation-related genes, and QRT-PCR was then carried out to verify the screened differentially expressed genes (DEGs). The results showed that, when stimulated by fetal bovine serum (10%) for 24 h or tumor necrosis factor-α (10 or 30 ng/ml) for 10 or 24 h, deletion of GATA4 C-terminus by p.S335X mutation in GATA4 enhanced the proliferation of RCSMC, without alteration of the migration capability. Twelve DEGs, including Fas, Hbegf, Itga5, Aimp1, Cxcl1, Il15, Il2rg, Il7, Tnfsf10, Il1r1, Irak1, and Tlr3, were screened and identified as phenotypic modulation-related genes. Our data might be beneficial for further exploration regarding the mechanisms of GATA4 p.S335X mutation on the phenotypic modulation of coronary VSMC.
Collapse
|
8
|
Ehrnthaller C, Braumüller S, Kellermann S, Gebhard F, Perl M, Huber-Lang M. Complement Factor C5a Inhibits Apoptosis of Neutrophils-A Mechanism in Polytrauma? J Clin Med 2021; 10:jcm10143157. [PMID: 34300323 PMCID: PMC8303460 DOI: 10.3390/jcm10143157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/02/2021] [Accepted: 07/15/2021] [Indexed: 01/03/2023] Open
Abstract
Life-threatening polytrauma results in early activation of the complement and apoptotic system, as well as leukocytes, ultimately leading to the clearance of damaged cells. However, little is known about interactions between the complement and apoptotic systems in PMN (polymorphonuclear neutrophils) after multiple injuries. PMN from polytrauma patients and healthy volunteers were obtained and assessed for apoptotic events along the post-traumatic time course. In vitro studies simulated complement activation by the exposure of PMN to C3a or C5a and addressed both the intrinsic and extrinsic apoptotic pathway. Specific blockade of the C5a-receptor 1 (C5aR1) on PMN was evaluated for efficacy to reverse complement-driven alterations. PMN from polytrauma patients exhibited significantly reduced apoptotic rates up to 10 days post trauma compared to healthy controls. Polytrauma-induced resistance was associated with significantly reduced Fas-ligand (FasL) and Fas-receptor (FasR) on PMN and in contrast, significantly enhanced FasL and FasR in serum. Simulation of systemic complement activation revealed for C5a, but not for C3a, a dose-dependent abrogation of PMN apoptosis in both intrinsic and extrinsic pathways. Furthermore, specific blockade of the C5aR1 reversed C5a-induced PMN resistance to apoptosis. The data suggest an important regulatory and putative mechanistic and therapeutic role of the C5a/C5aR1 interaction on PMN apoptosis after polytrauma.
Collapse
Affiliation(s)
- Christian Ehrnthaller
- Institute of Clinical and Experimental Trauma-Immunology (ITI), University of Ulm, 89081 Ulm, Germany; (S.B.); (S.K.)
- Department of Orthopedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), University Hospital, LMU Munich, 81377 Munich, Germany
- Correspondence: (C.E.); (M.H.-L.)
| | - Sonja Braumüller
- Institute of Clinical and Experimental Trauma-Immunology (ITI), University of Ulm, 89081 Ulm, Germany; (S.B.); (S.K.)
| | - Stephanie Kellermann
- Institute of Clinical and Experimental Trauma-Immunology (ITI), University of Ulm, 89081 Ulm, Germany; (S.B.); (S.K.)
| | - Florian Gebhard
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, Center of Surgery, University of Ulm, 89081 Ulm, Germany; (F.G.); (M.P.)
| | - Mario Perl
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, Center of Surgery, University of Ulm, 89081 Ulm, Germany; (F.G.); (M.P.)
- Department of Traumatology and Orthopaedic Surgery, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology (ITI), University of Ulm, 89081 Ulm, Germany; (S.B.); (S.K.)
- Correspondence: (C.E.); (M.H.-L.)
| |
Collapse
|
9
|
Suri GS, Kaur G, Jha CK, Tiwari M. Understanding idiopathic pulmonary fibrosis - Clinical features, molecular mechanism and therapies. Exp Gerontol 2021; 153:111473. [PMID: 34274426 DOI: 10.1016/j.exger.2021.111473] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 10/20/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic lung fibrosing disease with high prevalence that has a prognosis worse than many cancers. There has been a recent influx of new observations aimed at explaining the mechanisms responsible for the initiation and progression of pulmonary fibrosis. However, despite this, the pathogenesis of the disease is largely unclear. Recent progress has been made in the characterization of specific pathologic and clinical features that have enhanced the understanding of pathologically activated molecular pathways during the onset and progression of IPF. This review highlights several of the advances that have been made and focus on the pathobiology of IPF. The work also details the different factors that are responsible for the disposition of the disease - these may be internal factors such as cellular mechanisms and genetic alterations, or they may be external factors from the environment. The changes that primarily occur in epithelial cells and fibroblasts that lead to the activation of profibrotic pathways are discussed in depth. Finally, a complete repertoire of the treatment therapies that have been used in the past as well as future medications and therapies is provided.
Collapse
|
10
|
Berger RML, Weck JM, Kempe SM, Hill O, Liedl T, Rädler JO, Monzel C, Heuer-Jungemann A. Nanoscale FasL Organization on DNA Origami to Decipher Apoptosis Signal Activation in Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2101678. [PMID: 34057291 DOI: 10.1002/smll.202101678] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/13/2021] [Indexed: 05/27/2023]
Abstract
Cell signaling is initiated by characteristic protein patterns in the plasma membrane, but tools to decipher their molecular organization and activation are hitherto lacking. Among the well-known signaling pattern is the death inducing signaling complex with a predicted hexagonal receptor architecture. To probe this architecture, DNA origami-based nanoagents with nanometer precise arrangements of the death receptor ligand FasL are introduced and presented to cells. Mimicking different receptor geometries, these nanoagents act as signaling platforms inducing fastest time-to-death kinetics for hexagonal FasL arrangements with 10 nm inter-molecular spacing. Compared to naturally occurring soluble FasL, this trigger is faster and 100× more efficient. Nanoagents with different spacing, lower FasL number or higher coupling flexibility impede signaling. The results present DNA origami as versatile signaling scaffolds exhibiting unprecedented control over molecular number and geometry. They define molecular benchmarks in apoptosis signal initiation and constitute a new strategy to drive particular cell responses.
Collapse
Affiliation(s)
- Ricarda M L Berger
- Faculty of Physics and Center for NanoScience (CeNS), Ludwig-Maximilians-University, Geschwister-Scholl-Platz 1, 80539, Munich, Germany
| | - Johann M Weck
- Max Planck Institute of Biochemistry and Center for Nanoscience (CeNS), Ludwig-Maximilians-University, Am Klopferspitz 18, 82152, Martinsried, Germany
| | - Simon M Kempe
- Faculty of Physics and Center for NanoScience (CeNS), Ludwig-Maximilians-University, Geschwister-Scholl-Platz 1, 80539, Munich, Germany
| | - Oliver Hill
- Apogenix AG, University of Heidelberg, Im Neuenheimer Feld 584, 69120, Heidelberg, Germany
| | - Tim Liedl
- Faculty of Physics and Center for NanoScience (CeNS), Ludwig-Maximilians-University, Geschwister-Scholl-Platz 1, 80539, Munich, Germany
| | - Joachim O Rädler
- Faculty of Physics and Center for NanoScience (CeNS), Ludwig-Maximilians-University, Geschwister-Scholl-Platz 1, 80539, Munich, Germany
| | - Cornelia Monzel
- Experimental Medical Physics, Heinrich-Heine University, Universitätsstrasse 1, 40225, Düsseldorf, Germany
| | - Amelie Heuer-Jungemann
- Max Planck Institute of Biochemistry and Center for Nanoscience (CeNS), Ludwig-Maximilians-University, Am Klopferspitz 18, 82152, Martinsried, Germany
| |
Collapse
|
11
|
Shao R, Yang Y, Fan K, Wu X, Jiang R, Tang L, Li L, Shen Y, Liu G, Zhang L. REV-ERBα Agonist GSK4112 attenuates Fas-induced Acute Hepatic Damage in Mice. Int J Med Sci 2021; 18:3831-3838. [PMID: 34790059 PMCID: PMC8579287 DOI: 10.7150/ijms.52011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 10/07/2021] [Indexed: 12/04/2022] Open
Abstract
Fas-induced apoptosis is a central mechanism of hepatocyte damage during acute and chronic hepatic disorders. Increasing evidence suggests that circadian clock plays critical roles in the regulation of cell fates. In the present study, the potential significance of REV-ERBα, a core ingredient of circadian clock, in Fas-induced acute liver injury has been investigated. The anti-Fas antibody Jo2 was injected intraperitoneally in mice to induce acute liver injury and the REV-ERBα agonist GSK4112 was administered. The results indicated that treatment of GSK4112 decreased the level of plasma ALT and AST, attenuated the liver histological changes, and promoted the survival rate in Jo2-insulted mice. Treatment with GSK4112 also downregulated the activities of caspase-3 and caspase-8, suppressed hepatocyte apoptosis. In addition, treatment with GSK4112 decreased the level of Fas and enhanced the phosphorylation of Akt. In conclusion, treatment with GSK4112 alleviated Fas-induced apoptotic liver damage in mice, suggesting that REV-ERBα agonist might have potential value in pharmacological intervention of Fas-associated liver injury.
Collapse
Affiliation(s)
- Ruyue Shao
- Clinical Medical School, Chongqing Medical and Pharmaceutical College, Chongqing 401331, China.,Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing 401331, China
| | - Yongqiang Yang
- Department of Pathophysiology, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
| | - Kerui Fan
- Department of Pathophysiology, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
| | - Xicheng Wu
- Department of Pathophysiology, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
| | - Rong Jiang
- Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Li Tang
- Department of Pathophysiology, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
| | - Longjiang Li
- Department of Pathophysiology, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
| | - Yi Shen
- Department of Pathophysiology, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
| | - Gang Liu
- Department of Emergency, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - Li Zhang
- Department of Pathophysiology, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
12
|
Yusuf IO, Chen HM, Cheng PH, Chang CY, Tsai SJ, Chuang JI, Wu CC, Huang BM, Sun HS, Chen CM, Yang SH. FGF9 induces neurite outgrowth upon ERK signaling in knock-in striatal Huntington's disease cells. Life Sci 2020; 267:118952. [PMID: 33383048 DOI: 10.1016/j.lfs.2020.118952] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/07/2020] [Accepted: 12/16/2020] [Indexed: 12/17/2022]
Abstract
AIMS Huntington's disease (HD) is a neurodegenerative disease that causes deficits in neurite outgrowth, which suggests that enhancement of neurite outgrowth is a potential direction by which to improve HD. Our previous publications showed that fibroblast growth factor 9 (FGF9) provides anti-apoptosis and anti-oxidative functions in striatal cell models of HD through the extracellular signal-regulated kinases (ERK) pathway, and FGF9 also stimulates cytoskeletons to enhance neurite outgrowth via nuclear factor kappa B (NF-kB) signaling. In this study, we further demonstrate the importance of the ERK pathway for the neurite outgrowth induced by FGF9 in HD striatal models. MATERIALS AND METHODS FGF9 was treated with ERK (U0126) or NF-kB (BAY11-7082) inhibitors in STHdhQ7/Q7 and STHdhQ111/Q111 striatal knock-in cell lines to examine neurite outgrowth, cytoskeletal markers, and synaptic proteins via immunofluorescence staining and Western blotting. NF-kB activity was analyzed by NF-kB promoter reporter assay. KEY FINDINGS Here, we show that suppression of ERK signaling significantly inhibits FGF9-induced neurite outgrowth, cytoskeletal markers, and synaptic proteins in HD striatal cells. In addition, we also show suppression of ERK signaling significantly decreases FGF9-induced NF-kB activation, whereas suppression of NF-kB does not decrease FGF9-induced ERK signaling. These results suggest that FGF9 activates ERK signaling first, stimulates NF-kB upregulation, and then enhances neurite outgrowth in HD striatal cells. SIGNIFICANCE We elucidate the more detailed mechanisms of neurite outgrowth enhanced by FGF9 in these HD striatal cells. This study may provide insights into targeting neurite outgrowth for HD therapy.
Collapse
Affiliation(s)
- Issa Olakunle Yusuf
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Cheng Kung University and Academia Sinica, Taipei 11529, Taiwan; Institute of Clinical Medicine, College of Medicine, Taiwan; Department of Physiology, College of Medicine, Taiwan
| | - Hsiu-Mei Chen
- Department of Physiology, College of Medicine, Taiwan
| | | | - Chih-Yi Chang
- Department of Physiology, College of Medicine, Taiwan
| | - Shaw-Jenq Tsai
- Department of Physiology, College of Medicine, Taiwan; Institute of Basic Medical Sciences, Taiwan
| | - Jih-Ing Chuang
- Department of Physiology, College of Medicine, Taiwan; Institute of Basic Medical Sciences, Taiwan
| | - Chia-Ching Wu
- Institute of Basic Medical Sciences, Taiwan; Department of Cell Biology and Anatomy, Taiwan
| | - Bu-Miin Huang
- Institute of Basic Medical Sciences, Taiwan; Department of Cell Biology and Anatomy, Taiwan
| | - H Sunny Sun
- Institute of Basic Medical Sciences, Taiwan; Institute of Molecular Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Chuan-Mu Chen
- Department of Life Sciences, College of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan
| | - Shang-Hsun Yang
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Cheng Kung University and Academia Sinica, Taipei 11529, Taiwan; Department of Physiology, College of Medicine, Taiwan; Institute of Basic Medical Sciences, Taiwan.
| |
Collapse
|
13
|
Shao X, Liu L, Wei F, Liu Y, Wang F, Yi J, Sun L, Huang Y, Song Z, Yin W, Zhao H, Li Y. Fas and GIT1 signalling in the prefrontal cortex mediate behavioural sensitization to methamphetamine in mice. Brain Res Bull 2020; 164:361-371. [PMID: 32777257 DOI: 10.1016/j.brainresbull.2020.07.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 06/28/2020] [Accepted: 07/03/2020] [Indexed: 12/24/2022]
Abstract
PURPOSE Repeated methamphetamine (METH) administration in mice readily produces behavioural sensitization, but the underlying mechanisms remain elusive. The present research aimed to identify new targets affecting METH-induced behavioural sensitization. METHODS We first established a mouse model of METH-induced behavioural sensitization. To characterize the animal model, we performed behavioural tests at different stages of behavioural sensitization and simultaneously detected changes in several neurotransmitters in the prefrontal cortex (PFC). Next, we perfromed RNA sequencing (RNA-seq) to screen new targets, which were subsequently and verified by RT-PCR and western blot. Finally, we confirmed the roles of the new targets in METH-induced behavioural sensitization by injection of overexpressed lentiviruses and further detected related protein levels by western blot and histological changes by haematoxylin and eosin (HE) staining. RESULTS We successfully established a mouse model of METH-induced behavioural sensitization. The locomotor activities of the mice changed at different stages of sensitization, accompanied by changes in the levels of DA, 5-HT, GABA and glutamate. For RNA-seq analysis, we chose Fas as target, meanwhile, we chose GIT1 as target through literature. The detection of gene expression by RT-PCR indicated that METH-sensitized mice exhibited decreased levels of Fas, MEK1 and CREB and increased levels of Erk1/2 in the PFC. Western blot analysis revealed decreased Fas, GIT1, MEK1 and phosphorylated CREB levels and increased phosphorylated Erk1/2 levels in METH-sensitized mice. Injection of Fas and GIT1 injection showed that overexpression of Fas and GIT1 inhibited the induction of METH sensitization and reversed the changes in neurotransmitter levels and related protein levels, including MEK1, phosphorylated CREB and phosphorylated Erk1/2, in METH-sensitized mice. Overexpression of Fas and GIT1 also reduced histological lesions induced by METH. CONCLUSION The findings indicated that the development of behavioural sensitization to METH may be mediated by Fas and GIT1 through the MEK1-Erk1/2-CREB pathway.
Collapse
Affiliation(s)
- Xiaotong Shao
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130024, China
| | - Lei Liu
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130024, China
| | - Fuyao Wei
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130024, China
| | - Yucui Liu
- Research Center of Agriculture and Medicine Gene Engineering of Ministry of Education, Northeast Normal University, Changchun, 130024, China
| | - Fei Wang
- Research Center of Agriculture and Medicine Gene Engineering of Ministry of Education, Northeast Normal University, Changchun, 130024, China
| | - Jingwen Yi
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130024, China
| | - Luguo Sun
- Research Center of Agriculture and Medicine Gene Engineering of Ministry of Education, Northeast Normal University, Changchun, 130024, China
| | - Yanxin Huang
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130024, China
| | - Zhenbo Song
- Research Center of Agriculture and Medicine Gene Engineering of Ministry of Education, Northeast Normal University, Changchun, 130024, China
| | - Wu Yin
- Research Center of Agriculture and Medicine Gene Engineering of Ministry of Education, Northeast Normal University, Changchun, 130024, China.
| | - Huiying Zhao
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yunxin Li
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130024, China
| |
Collapse
|
14
|
Antidiabetic Effects of Arginyl-Fructosyl-Glucose, a Nonsaponin Fraction from Ginseng Processing in Streptozotocin-Induced Type 2 Diabetic Mice through Regulating the PI3K/AKT/GSK-3 β and Bcl-2/Bax Signaling Pathways. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:3707904. [PMID: 32714403 PMCID: PMC7352147 DOI: 10.1155/2020/3707904] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 04/06/2020] [Indexed: 01/21/2023]
Abstract
Streptozotocin- (STZ-) induced type 2 diabetes mellitus (T2DM) caused insulin secretion disorder and hyperglycemia, further causing tissue and organ damage. In recent years, studies on ginseng (Panax ginseng C. A. Meyer) and its saponins (Ginsenosides) have proved to possess antidiabetic pharmacological activities, but the mechanism of nonsaponins on STZ-induced T2DM is still unclear. Arginyl-fructosyl-glucose (AFG) is a representative nonsaponin component produced in the processing of red ginseng. The present study was designed to assess the possible healing consequence of AFG on STZ-induced T2DM in mice and also to explore its fundamental molecular contrivances. T2DM-related indexes, fasting blood glucose levels, and body weight, histological changes, biochemical considerations, biomarkers, the mRNA countenance intensities of inflammatory facts, and variations in correlated protein manifestation in adipose tissue and liver tissue were calculated. Consequences specified that AFG usage successfully amends STZ-induced insulin conflict and liver grievance in T2DM. Systematically, AFG action diminished STZ-induced oxidative stress and inflammatory responses in the liver. In addition, we demonstrated that AFG also attenuates apoptosis and insulin secretion disorders in T2DM by adjusting the PI3K/AKT/GSK3β signaling pathway. At the end, these discoveries recommend that AFG averts the development of T2DM through numerous types of machinery and proposes that AFG can also be used in order to treat T2DM in the future.
Collapse
|
15
|
Jaufmann J, Lelis FJN, Teschner AC, Fromm K, Rieber N, Hartl D, Beer-Hammer S. Human monocytic myeloid-derived suppressor cells impair B-cell phenotype and function in vitro. Eur J Immunol 2019; 50:33-47. [PMID: 31557313 DOI: 10.1002/eji.201948240] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/16/2019] [Accepted: 09/25/2019] [Indexed: 01/06/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are key regulators of immunity that initially have been defined by their ability to potently suppress T-cell responses. Recent studies collectively demonstrate that the suppressive activity of MDSCs is not limited to T cells, but rather affects a broad range of immune cell subsets. However, relatively few studies have assessed the impact of MDSCs on B cells, particularly in the human context. Here, we report that human monocytic MDSCs (M-MDSCs) significantly interfere with human B-cell proliferation and function in vitro. We further show that the inhibition occurs independent of direct cell-contact and involves the expression of suppressive mediators such as indoleamine 2, 3-dioxygenase (IDO), arginase-1 (Arg1), and nitric oxide (NO). In addition, our studies demonstrate that the suppression of B cells by M-MDSCs is paralleled by a skewing in B-cell phenotype and gene expression signatures. M-MDSCs induced the downregulation of key surface markers on activated B cells, including IgM, HLA-DR, CD80, CD86, TACI, and CD95. Concurrently, M-MDSCs but not conventional monocytes elicited alterations in the transcription of genes involved in apoptosis induction, class-switch regulation, and B-cell differentiation and function. In summary, this study expands our understanding of the regulatory role of M-MDSCs for human B-cell responses.
Collapse
Affiliation(s)
- Jennifer Jaufmann
- Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology and ICePhA, University of Tuebingen, Tuebingen, Germany.,Children's Hospital and Interdisciplinary Center for Infectious Diseases, University of Tuebingen, Tuebingen, Germany
| | - Felipe J N Lelis
- Children's Hospital and Interdisciplinary Center for Infectious Diseases, University of Tuebingen, Tuebingen, Germany.,Department of Medicine, Division of Rheumatology, Immunology and Allergy Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, MA, USA
| | - Annkathrin C Teschner
- Children's Hospital and Interdisciplinary Center for Infectious Diseases, University of Tuebingen, Tuebingen, Germany
| | - Katja Fromm
- Children's Hospital and Interdisciplinary Center for Infectious Diseases, University of Tuebingen, Tuebingen, Germany.,Biozentrum, University of Basel, Infection Biology, Basel, Switzerland
| | - Nikolaus Rieber
- Children's Hospital and Interdisciplinary Center for Infectious Diseases, University of Tuebingen, Tuebingen, Germany.,Department of Pediatrics, Kinderklinik Muenchen Schwabing, Muenchen Klinik und Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Dominik Hartl
- Children's Hospital and Interdisciplinary Center for Infectious Diseases, University of Tuebingen, Tuebingen, Germany
| | - Sandra Beer-Hammer
- Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology and ICePhA, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
16
|
Savari F, Badavi M, Rezaie A, Gharib-Naseri MK, Mard SA. Evaluation of the therapeutic potential effect of Fas receptor gene knockdown in experimental model of non-alcoholic steatohepatitis. Free Radic Res 2019; 53:486-496. [PMID: 31010354 DOI: 10.1080/10715762.2019.1608982] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Aim: Stimulation of Fas death receptor is introduced as a major cause of non-alcoholic steatohepatitis (NASH) progression through suppression of cell viability. Therefore, the blocking of death pathways is hypothesised to be express new approaches to NASH therapy. For this purpose, current experiment applied synthetic small interference RNA (SiRNA) to trigger Fas death receptor and to show its potential therapeutic role in designed NASH model. Methods: Male mice were placed on a western diet (WD) for 8 weeks and exposed to cigarette smoke during the last 4 weeks of feeding to induce NASH model. In the next step, Fas SiRNA was injected to mice aiming to examine specific Fas gene silencing, after 8 weeks. As a control, mice received scrambled SiRNA. Reversible possibility of disease was examined by 3 weeks of recovery. Results: Analysis of data is accompanied with the significant histopathological changes (steatosis, ballooning and inflammation), increased lipid profile and hepatic enzyme activities (AST, ALT, ALP) plus TBARS as well as decreased antioxidants levels in NASH model. Upon Fas-SiRNA injection, almost all measured parameters of NASH such as overexpression of Fas receptor, caspase3, NF-kB genes and marked increase of hepatic TNF-α were significantly restored and were remained nearly unchanged following recovery liking as scrambled groups. Conclusions: The suppression of Fas receptor signalling subsequent RNAi therapy may represent an applicable strategy to decline hepatocyte damages and so NASH progression in mice.
Collapse
Affiliation(s)
- Feryal Savari
- a Physiology Research Center (PRC), Department of Physiology, School of Medicine , Ahvaz Jundishapur University of Medical Sciences , Ahvaz , Iran
| | - Mohammad Badavi
- a Physiology Research Center (PRC), Department of Physiology, School of Medicine , Ahvaz Jundishapur University of Medical Sciences , Ahvaz , Iran
| | - Anahita Rezaie
- b Department of Pathobiology, School of Veterinary Medicine , Shahid Chamran University of Ahvaz , Ahvaz , Iran
| | - Mohammad Kazem Gharib-Naseri
- a Physiology Research Center (PRC), Department of Physiology, School of Medicine , Ahvaz Jundishapur University of Medical Sciences , Ahvaz , Iran
| | - Seyyed Ali Mard
- c Persian Gulf's Physiology Research Center (PRC), Alimentary Tract Research Center, Department of Physiology, School of Medicine , Ahvaz Jundishapur University of Medical Sciences , Ahvaz , Iran
| |
Collapse
|
17
|
Brennan K, Lyons C, Fernandes P, Doyle S, Houston A, Brint E. Engagement of Fas differentially regulates the production of LPS-induced proinflammatory cytokines and type I interferons. FEBS J 2018; 286:523-535. [PMID: 30536547 DOI: 10.1111/febs.14727] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 10/22/2018] [Accepted: 12/07/2018] [Indexed: 12/28/2022]
Abstract
Fas (CD95) signalling is best known for its role in apoptosis, however, recent reports have shown it to be involved in other cellular responses as well, including inflammation. Fas and its adaptor protein FADD are known to negatively regulate LPS-induced proinflammatory responses, but their role in LPS-induced type I interferon production is unknown. Here, we demonstrate that Fas engagement on macrophages, using an agonistic Fas antibody CH11, augments LPS-induced NF-κB responses, causing increased production of TNFα, IL-8, IL-6 and IL-12. Conversely, costimulation with both LPS and CH11 causes a significant reduction in the level of interferon-beta (IFNβ) production. This differential effect involves the Fas adaptor FADD because while LPS-induced IL-6 production increased in FADD-/- murine embryonic fibroblasts, LPS-induced IFNβ production was significantly reduced in these cells. Overexpression of a dominant negative form of FADD (FADD-DD) inhibits LPS-induced IFNβ luciferase but not LPS-induced NF-κB luciferase. In contrast, overexpression of full-length FADD inhibited LPS-induced NF-κB luciferase activation but was seen to augment LPS-induced IFNβ luciferase. Moreover, FADD-DD inhibits TRIF-, TRAM-, IKKε-, TBK-1- and TRAF3-induced IFNβ luciferase production, with coimmunoprecipitation experiments demonstrating an interaction between FADD and TRIF. These data identify FADD as a novel component of the noncanonical Toll-like receptor 4/IFNβ signalling pathway and demonstrate that both Fas and its adaptor FADD can differentially regulate the production of LPS-induced proinflammatory cytokines and type I interferons.
Collapse
Affiliation(s)
- Kiva Brennan
- Department of Clinical Medicine School of Medicine, Trinity College Dublin, Ireland.,The National Children's Research Centre, Dublin, Ireland
| | | | | | - Sarah Doyle
- Department of Clinical Medicine School of Medicine, Trinity College Dublin, Ireland.,The National Children's Research Centre, Dublin, Ireland
| | - Aileen Houston
- Department of Medicine, University College Cork, Ireland.,APC Microbiome Ireland, University College Cork, Ireland
| | - Elizabeth Brint
- Department of Pathology, University College Cork, Ireland.,APC Microbiome Ireland, University College Cork, Ireland
| |
Collapse
|
18
|
Mande P, Zirak B, Ko WC, Taravati K, Bride KL, Brodeur TY, Deng A, Dresser K, Jiang Z, Ettinger R, Fitzgerald KA, Rosenblum MD, Harris JE, Marshak-Rothstein A. Fas ligand promotes an inducible TLR-dependent model of cutaneous lupus-like inflammation. J Clin Invest 2018; 128:2966-2978. [PMID: 29889098 PMCID: PMC6025993 DOI: 10.1172/jci98219] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 04/17/2018] [Indexed: 02/06/2023] Open
Abstract
Toll-like receptors TLR7 and TLR9 are both implicated in the activation of autoreactive B cells and other cell types associated with systemic lupus erythematosus (SLE) pathogenesis. However, Tlr9-/- autoimmune-prone strains paradoxically develop more severe disease. We have now leveraged the negative regulatory role of TLR9 to develop an inducible rapid-onset murine model of systemic autoimmunity that depends on T cell detection of a membrane-bound OVA fusion protein expressed by MHC class II+ cells, expression of TLR7, expression of the type I IFN receptor, and loss of expression of TLR9. These mice are distinguished by a high frequency of OVA-specific Tbet+, IFN-γ+, and FasL-expressing Th1 cells as well as autoantibody-producing B cells. Unexpectedly, contrary to what occurs in most models of SLE, they also developed skin lesions that are very similar to those of human cutaneous lupus erythematosus (CLE) as far as clinical appearance, histological changes, and gene expression. FasL was a key effector mechanism in the skin, as the transfer of FasL-deficient DO11gld T cells completely failed to elicit overt skin lesions. FasL was also upregulated in human CLE biopsies. Overall, our model provides a relevant system for exploring the pathophysiology of CLE as well as the negative regulatory role of TLR9.
Collapse
Affiliation(s)
- Purvi Mande
- Department of Medicine, University of Massachusetts School of Medicine, Worcester, Massachusetts, USA
| | - Bahar Zirak
- Department of Dermatology, UCSF, San Francisco, California, USA
| | - Wei-Che Ko
- Department of Dermatology, University of Massachusetts School of Medicine, Worcester, Massachusetts, USA
| | - Keyon Taravati
- Department of Dermatology, UCSF, San Francisco, California, USA
| | - Karen L Bride
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Tia Y Brodeur
- Department of Medicine, University of Massachusetts School of Medicine, Worcester, Massachusetts, USA
| | - April Deng
- Department of Dermatology, University of Massachusetts School of Medicine, Worcester, Massachusetts, USA
| | - Karen Dresser
- Department of Dermatology, University of Massachusetts School of Medicine, Worcester, Massachusetts, USA
| | - Zhaozhao Jiang
- Department of Medicine, University of Massachusetts School of Medicine, Worcester, Massachusetts, USA
| | - Rachel Ettinger
- Respiratory, Autoimmunity, and Inflammation Department, MedImmune, Gaithersburg, Maryland, USA
| | - Katherine A Fitzgerald
- Department of Medicine, University of Massachusetts School of Medicine, Worcester, Massachusetts, USA
| | | | - John E Harris
- Department of Dermatology, University of Massachusetts School of Medicine, Worcester, Massachusetts, USA
| | - Ann Marshak-Rothstein
- Department of Medicine, University of Massachusetts School of Medicine, Worcester, Massachusetts, USA.,Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
19
|
Chen M, Cai F, Zha D, Wang X, Zhang W, He Y, Huang Q, Zhuang H, Hua ZC. Astragalin-induced cell death is caspase-dependent and enhances the susceptibility of lung cancer cells to tumor necrosis factor by inhibiting the NF-кB pathway. Oncotarget 2018; 8:26941-26958. [PMID: 28199969 PMCID: PMC5432309 DOI: 10.18632/oncotarget.15264] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 01/22/2017] [Indexed: 12/15/2022] Open
Abstract
Flavonoids are naturally occurring polyphenolic compounds and are among the most promising anticancer agents. Here, we demonstrate that the flavonoid astragalin (AG), also known as kaempferol-3-O-β-D-glucoside, induces cell death. This was prevented by the caspase inhibitors z-DEVD-FMK and z-LEHD-FMK. AG-induced cell death was associated with an increase in the Bax:Bcl-2 ratio and amplified by the inhibition of extracellular signal-regulated kinase (ERK)-1/2 and Akt signaling. Meanwhile, AG suppressed LPS-induced NF-κB activation. Additional studies revealed that AG inhibited tumor necrosis factor-alpha (TNFα)-induced NF-κB activity. AG also potentiated TNFα-induced apoptosis in A549 cells. Furthermore, using a mouse xenograft model, we demonstrated that AG suppressed tumor growth and induced cancer cell apoptosis in vivo. Taken together, these results suggest that AG may be a promising cancer therapeutic drug that warrants further investigation into its potential clinical applications.
Collapse
Affiliation(s)
- Minghui Chen
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.,State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau
| | - Fangfang Cai
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Daolong Zha
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Xueshi Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Wenjing Zhang
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau
| | - Yan He
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau
| | - Qilai Huang
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau.,Changzhou High-Tech Research Institute of Nanjing University and Target Pharma Laboratory, Changzhou, Jiangsu, China
| | - Hongqin Zhuang
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.,Changzhou High-Tech Research Institute of Nanjing University and Target Pharma Laboratory, Changzhou, Jiangsu, China
| | - Zi-Chun Hua
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.,State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau.,College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
20
|
Knoll G, Bittner S, Kurz M, Jantsch J, Ehrenschwender M. Hypoxia regulates TRAIL sensitivity of colorectal cancer cells through mitochondrial autophagy. Oncotarget 2018; 7:41488-41504. [PMID: 27166192 PMCID: PMC5173074 DOI: 10.18632/oncotarget.9206] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 04/24/2016] [Indexed: 11/25/2022] Open
Abstract
The capacity of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) to selectively induce cell death in malignant cells triggered numerous attempts for therapeutic exploitation. In clinical trials, however, TRAIL did not live up to the expectations, as tumors exhibit high rates of TRAIL resistance in vivo. Response to anti-cancer therapy is determined not only by cancer cell intrinsic factors (e.g. oncogenic mutations), but also modulated by extrinsic factors such as the hypoxic tumor microenvironment.Here, we address the effect of hypoxia on pro-apoptotic TRAIL signaling in colorectal cancer cells. We show that oxygen levels modulate susceptibility to TRAIL-induced cell death, which is severely impaired under hypoxia (0.5% O2). Mechanistically, this is attributable to hypoxia-induced mitochondrial autophagy. Loss of mitochondria under hypoxia restricts the availability of mitochondria-derived pro-apoptotic molecules such as second mitochondria-derived activator of caspase (SMAC), thereby disrupting amplification of the apoptotic signal emanating from the TRAIL death receptors and efficiently blocking cell death in type-II cells. Moreover, we identify strategies to overcome TRAIL resistance in low oxygen environments. Counteracting hypoxia-induced loss of endogenous SMAC by exogenous substitution of SMAC mimetics fully restores TRAIL sensitivity in colorectal cancer cells. Alternatively, enforcing a mitochondria-independent type-I mode of cell death by targeting the type-II phenotype gatekeeper X-linked inhibitor of apoptosis protein (XIAP) is equally effective.Together, our results indicate that tumor hypoxia impairs TRAIL efficacy but this limitation can be overcome by combining TRAIL with SMAC mimetics or XIAP-targeting drugs. Our findings may help to exploit the potential of TRAIL in cancer therapy.
Collapse
Affiliation(s)
- Gertrud Knoll
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Sebastian Bittner
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Maria Kurz
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Jonathan Jantsch
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Martin Ehrenschwender
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
21
|
Balomenos D, Shokri R, Daszkiewicz L, Vázquez-Mateo C, Martínez-A C. On How Fas Apoptosis-Independent Pathways Drive T Cell Hyperproliferation and Lymphadenopathy in lpr Mice. Front Immunol 2017; 8:237. [PMID: 28344578 PMCID: PMC5344898 DOI: 10.3389/fimmu.2017.00237] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 02/20/2017] [Indexed: 01/20/2023] Open
Abstract
Fas induces massive apoptosis in T cells after repeated in vitro T cell receptor (TCR) stimulation and is critical for lymphocyte homeostasis in Fas-deficient (lpr) mice. Although the in vitro Fas apoptotic mechanism has been defined, there is a large conceptual gap between this in vitro phenomenon and the pathway that leads to in vivo development of lymphadenopathy and autoimmunity. A striking abnormality in lpr mice is the excessive proliferation of CD4+ and CD8+ T cells, and more so of the double-negative TCR+CD4−CD8−B220+ T cells. The basis of lpr T cell hyperproliferation remains elusive, as it cannot be explained by Fas-deficient apoptosis. T cell-directed p21 overexpression reduces hyperactivation/hyperproliferation of all lpr T cell subtypes and lymphadenopathy in lpr mice. p21 controls expansion of repeatedly stimulated T cells without affecting apoptosis. These results confirm a direct link between hyperactivation/hyperproliferation, autoreactivity, and lymphadenopathy in lpr mice and, with earlier studies, suggest that Fas apoptosis-independent pathways control lpr T cell hyperproliferation. lpr T cell hyperproliferation could be an indirect result of the defective apoptosis of repeatedly stimulated lpr T cells. Nonetheless, in this perspective, we argue for an alternative setting, in which lack of Fas would directly cause lpr T cell hyperactivation/hyperproliferation in vivo. We propose that Fas/Fas ligand (FasL) acts as an activation inhibitor of recurrently stimulated T cells, and that its disruption causes overexpansion of T cells in lpr mice. Research to define the underlying mechanism of this Fas/FasL effect could resolve the phenotype of lpr mice and lead to therapeutics for related human syndromes.
Collapse
Affiliation(s)
- Dimitrios Balomenos
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), UAM Campus de Cantoblanco , Madrid , Spain
| | - Rahman Shokri
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), UAM Campus de Cantoblanco , Madrid , Spain
| | - Lidia Daszkiewicz
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), UAM Campus de Cantoblanco , Madrid , Spain
| | - Cristina Vázquez-Mateo
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), UAM Campus de Cantoblanco , Madrid , Spain
| | - Carlos Martínez-A
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), UAM Campus de Cantoblanco , Madrid , Spain
| |
Collapse
|
22
|
Nickerson KM, Wang Y, Bastacky S, Shlomchik MJ. Toll-like receptor 9 suppresses lupus disease in Fas-sufficient MRL Mice. PLoS One 2017; 12:e0173471. [PMID: 28278279 PMCID: PMC5344451 DOI: 10.1371/journal.pone.0173471] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 02/22/2017] [Indexed: 12/26/2022] Open
Abstract
Genetic deficiency in TLR9 accelerates pathogenesis in the spontaneous polygenic MRL.Faslpr murine model of systemic lupus erythematosus, despite the absence of anti-nucleosome autoantibodies. However, it could be argued that this result was dependent on Fas-deficiency rather than lupus-promoting genes in the MRL genetic background. Here we report the effects of TLR9 deficiency on autoimmune disease independent of the lpr mutation in Fas by characterizing Tlr9-/- and Tlr9+/+ mice on the Fas-intact MRL/+ genetic background. By 30 weeks of age, Tlr9-deficient MRL/+ had more severe renal disease, increased T cell activation, and higher titers of anti-Sm and anti-RNA autoantibodies than Tlr9-intact animals, as had been the case in the MRL.Faslpr model. In addition, Tlr9-deficient MRL/+ mice had increased numbers of germinal center phenotype B cells and an increase in splenic neutrophils and conventional dendritic cell populations. Thus, the disease accelerating effects of Tlr9 deficiency are separable from those mediated by the Fas mutation in the lupus-prone MRL genetic background. Nonetheless, disease acceleration in Tlr9-deficient MRL/+ mice was phenotypically distinct from that in Fas-deficient counterparts, which has important implications.
Collapse
Affiliation(s)
- Kevin M. Nickerson
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| | - Yujuan Wang
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Sheldon Bastacky
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Mark J. Shlomchik
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
23
|
Lund N, Gränsbo K, Wernersson C, Melander O. Cardiometabolic biomarkers are predictors of readmission and death in patients hospitalized for acute dyspnea. Am J Emerg Med 2016; 35:610-614. [PMID: 28062207 PMCID: PMC5754318 DOI: 10.1016/j.ajem.2016.12.048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 12/12/2016] [Accepted: 12/16/2016] [Indexed: 12/20/2022] Open
Abstract
Background Acute dyspnea affects a large heterogeneous patient group with high mortality and readmission rates. Purpose To investigate if cardiometabolic biomarkers and clinical characteristics predict readmission and death in patients hospitalized for acute dyspnea. Methods 65 dyspnea patients at a general internal medicine ward were followed for six months. The combined endpoint was readmission or death. Measurements and results Cardiometabolic biomarkers at admission were related to the endpoint in Cox proportional hazard models (adjusted for sex, age, oxygen saturation, respiratory rate and C-reactive protein (CRP)). The biomarkers tissue-type plasminogen activator (tPA), prolactin (PRL), tumor necrosis factor receptor superfamily member 6 (FAS) and C-C motif chemokine 3 (CCL3) were independently and significantly related to the endpoint and combined into a biomarker risk score (BRS). Each SD increment of the BRS conferred a hazard ratio (HR) of 2.13 (1.39–3.27) P = 0.001. The top vs bottom tertile of the BRS conferred a HR of 4.75 (1.93–11.68) P = 0.001. Dyspnea severity was also associated with worse outcome, HR = 3.43 (1.28–9.20) P = 0.014. However, when mutually adjusted the BRS remained significant (P = 0.004) whereas dyspnea severity was not. The BRS was related to the endpoint among patients with mild to moderate dyspnea (P = 0.016) but not among those with severe dyspnea. Conclusion A score of tPA, PRL, FAS and CCL3 predicts 6-month death and readmission in patients hospitalized for acute dyspnea and may prove useful to optimize length of stay and follow-up. Although the BRS outweighs dyspnea severity in prediction of the endpoint, its prognostic role is strongest in mild-moderate dyspnea.
Collapse
Affiliation(s)
- Nathalie Lund
- Skåne University Hospital Malmö, Clinical Research Centre, Malmoe, Sweden.
| | - Klas Gränsbo
- Skåne University Hospital Malmö, Clinical Research Centre CRC, Malmoe, Sweden
| | - Camilla Wernersson
- Skåne University Hospital Malmö, Clinical Research Centre CRC, Malmoe, Sweden
| | - Olle Melander
- Skåne University Hospital Malmö, Clinical Research Centre CRC, Malmoe, Sweden
| |
Collapse
|
24
|
MiR-106a: Promising biomarker for cancer. Bioorg Med Chem Lett 2016; 26:5373-5377. [PMID: 27780637 DOI: 10.1016/j.bmcl.2016.10.042] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 09/23/2016] [Accepted: 10/13/2016] [Indexed: 12/19/2022]
Abstract
MicroRNAs (miRNAs), which are characterized by highly conserved and small non-coding RNAs, have been a hot spot regarding biological processes such as cellular proliferation, apoptosis and metabolism as well as cellular differentiation, signal transduction and carcinogenesis. MiRNA-106a (miR-106a), a member of the miR-17 family, has been validated to be aberrantly regulated in the diversity of tumors. The purpose of this review is supposed to deliver an intricate overview of miR-106a, including its role in cell proliferation, apoptosis, cell cycle, invasion and metastasis, involvement in drug resistance as well as its interactions with the target proteins and signaling pathways involved.
Collapse
|
25
|
Im J, Kim K, Hergert P, Nho RS. Idiopathic pulmonary fibrosis fibroblasts become resistant to Fas ligand-dependent apoptosis via the alteration of decoy receptor 3. J Pathol 2016; 240:25-37. [PMID: 27218286 DOI: 10.1002/path.4749] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 05/03/2016] [Accepted: 05/09/2016] [Indexed: 11/06/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is an irreversible lethal lung disease with an unknown etiology. IPF patients' lung fibroblasts express inappropriately high Akt activity, protecting them in response to an apoptosis-inducing type I collagen matrix. FasL, a ligand for Fas, is known to be increased in the lung tissues of patients with IPF, implicated with the progression of IPF. Expression of Decoy Receptor3 (DcR3), which binds to FasL, thereby subsequently suppressing the FasL-Fas-dependent apoptotic pathway, is frequently altered in various human disease. However, the role of DcR3 in IPF fibroblasts in regulating their viability has not been examined. We found that enhanced DcR3 expression exists in the majority of IPF fibroblasts on collagen matrices, resulting in the protection of IPF fibroblasts from FasL-induced apoptosis. Abnormally high Akt activity suppresses GSK-3β function, thereby accumulating the nuclear factor of activated T-cells cytoplasmic 1 (NFATc1) in the nucleus, increasing DcR3 expression in IPF fibroblasts. This alteration protects IPF cells from FasL-induced apoptosis on collagen. However, the inhibition of Akt or NFATc1 decreases DcR3 mRNA and protein levels, which sensitizes IPF fibroblasts to FasL-mediated apoptosis. Furthermore, enhanced DcR3 and NFATc1 expression is mainly present in myofibroblasts in the fibroblastic foci of lung tissues derived from IPF patients. Our results showed that when IPF cells interact with collagen matrix, aberrantly activated Akt increases DcR3 expression via GSK-3β-NFATc1 and protects IPF cells from the FasL-dependent apoptotic pathway. These findings suggest that the inhibition of DcR3 function may be an effective approach for sensitizing IPF fibroblasts in response to FasL, limiting the progression of lung fibrosis. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jintaek Im
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Kyutae Kim
- College of Biological Science, University of Minnesota, St. Paul, MN, USA
| | - Polla Hergert
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA.,Lung Morphology Research Core Department, University of Minnesota, Minneapolis, MN, USA
| | | |
Collapse
|
26
|
Cytotoxicity of polymethyl methacrylate cement on primary cultured metastatic spinal cells. Mol Cell Toxicol 2016. [DOI: 10.1007/s13273-016-0016-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
27
|
Klein S, Dieterich LC, Mathelier A, Chong C, Sliwa-Primorac A, Hong YK, Shin JW, Lizio M, Itoh M, Kawaji H, Lassmann T, Daub CO, Arner E, Carninci P, Hayashizaki Y, Forrest ARR, Wasserman WW, Detmar M. DeepCAGE transcriptomics identify HOXD10 as a transcription factor regulating lymphatic endothelial responses to VEGF-C. J Cell Sci 2016; 129:2573-85. [PMID: 27199372 DOI: 10.1242/jcs.186767] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 05/11/2016] [Indexed: 01/15/2023] Open
Abstract
Lymphangiogenesis plays a crucial role during development, in cancer metastasis and in inflammation. Activation of VEGFR-3 (also known as FLT4) by VEGF-C is one of the main drivers of lymphangiogenesis, but the transcriptional events downstream of VEGFR-3 activation are largely unknown. Recently, we identified a wave of immediate early transcription factors that are upregulated in human lymphatic endothelial cells (LECs) within the first 30 to 80 min after VEGFR-3 activation. Expression of these transcription factors must be regulated by additional pre-existing transcription factors that are rapidly activated by VEGFR-3 signaling. Using transcription factor activity analysis, we identified the homeobox transcription factor HOXD10 to be specifically activated at early time points after VEGFR-3 stimulation, and to regulate expression of immediate early transcription factors, including NR4A1. Gain- and loss-of-function studies revealed that HOXD10 is involved in LECs migration and formation of cord-like structures. Furthermore, HOXD10 regulates expression of VE-cadherin, claudin-5 and NOS3 (also known as e-NOS), and promotes lymphatic endothelial permeability. Taken together, these results reveal an important and unanticipated role of HOXD10 in the regulation of VEGFR-3 signaling in lymphatic endothelial cells, and in the control of lymphangiogenesis and permeability.
Collapse
Affiliation(s)
- Sarah Klein
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zurich, Zurich 8093, Switzerland
| | - Lothar C Dieterich
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zurich, Zurich 8093, Switzerland
| | - Anthony Mathelier
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University British Columbia, Vancouver, British Columbia, Canada V5Z 4H4
| | - Chloé Chong
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zurich, Zurich 8093, Switzerland
| | - Adriana Sliwa-Primorac
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zurich, Zurich 8093, Switzerland
| | - Young-Kwon Hong
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Jay W Shin
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Yokohama, Kanagawa 230-0045, Japan
| | - Marina Lizio
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Yokohama, Kanagawa 230-0045, Japan
| | - Masayoshi Itoh
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Yokohama, Kanagawa 230-0045, Japan
| | - Hideya Kawaji
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Yokohama, Kanagawa 230-0045, Japan
| | - Timo Lassmann
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Yokohama, Kanagawa 230-0045, Japan Telethon Kids Institute, The University of Western Australia, Subiaco, Western Australia 6008, Australia
| | - Carsten O Daub
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Yokohama, Kanagawa 230-0045, Japan
| | - Erik Arner
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Yokohama, Kanagawa 230-0045, Japan
| | | | - Piero Carninci
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Yokohama, Kanagawa 230-0045, Japan
| | - Yoshihide Hayashizaki
- RIKEN Preventive Medicine and Diagnosis Innovation Program, Wako, Saitama 351-0198, Japan
| | - Alistair R R Forrest
- RIKEN Center for Life Science Technologies, Division of Genomic Technologies, Yokohama, Kanagawa 230-0045, Japan Cancer and Cell Biology Division, Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, the University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Wyeth W Wasserman
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University British Columbia, Vancouver, British Columbia, Canada V5Z 4H4
| | - Michael Detmar
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zurich, Zurich 8093, Switzerland
| |
Collapse
|
28
|
Abstract
While mesenchymal stem cells (MSCs) are rapidly cleared from the body following systemic transplantation, their therapeutic benefits typically persist. In this issue, Liu et al. (2015) reveal that the ability of transplanted MSCs to alleviate osteoporosis in systemic lupus erythematosus is maintained through epigenetic changes conferred by secretory action of the MSCs.
Collapse
Affiliation(s)
- Kelvin S Ng
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA; Department of Medicine, Division of Biomedical Engineering, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Thomas M Kuncewicz
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA; Department of Medicine, Division of Biomedical Engineering, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Jeffrey M Karp
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA; Department of Medicine, Division of Biomedical Engineering, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| |
Collapse
|
29
|
Jiang G, Wang Y, Yun J, Hajrasouliha AR, Zhao Y, Sun D, Kaplan HJ, Shao H. HMGB1 release triggered by the interaction of live retinal cells and uveitogenic T cells is Fas/FasL activation-dependent. J Neuroinflammation 2015; 12:179. [PMID: 26394985 PMCID: PMC4579830 DOI: 10.1186/s12974-015-0389-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 09/02/2015] [Indexed: 11/29/2022] Open
Abstract
Background It is not clear how invading autoreactive T cells initiate the pathogenic process inside the diseased organ in T cell-mediated organ-specific autoimmune disease. In experimental autoimmune uveitis (EAU) induced by adoptive transfer of interphotoreceptor retinoid-binding protein (IRBP)-specific T cells in mice, we have previously reported that intraocular inflammation was initiated by infiltrating IRBP-specific T cells that directly interacted with retinal cells and resulted in the active release of high mobility group box 1 (HMGB1), an important member of damage associate molecular patterns (DAMPs). Furthermore, blockade of HMGB1 in our murine model reduced intraocular inflammation via suppression of IRBP-specific T cell functions. These results have demonstrated that HMGB1 is an early and critical mediator of induction of intraocular inflammation. The present study identified the cell surface molecule that triggers HMGB1 secretion. Methods Retinal explants from Fas-deficient (Faslpr) and wild-type (Wt) C57BL/6 (B6) mice were cultured with activated IRBP 1–20 peptide-specific T cells or with a Fas-activating antibody (Jo2), and then the level of HMGB1 in culture supernatants were detected by ELISA. In addition, released HMGB1 was examined in the eye of Faslpr and Wt mice after IRBP-specific T cell transfer. Uveitis was evaluated in the IRBP-specific T cell transferred Faslpr mice after recombinant HMGB1 was restored within the eye and in the IRBP-specific T cell transferred Wt mice after they were treated with a Fas antagonist (Met12). Results In contrast to retinal explants from Wt mice, those from Faslpr mice did not release HMGB1 after exposure to IRBP-specific T cells or to Jo2. The release of HMGB1 by Wt retinal explants was suppressed by Met 12. Moreover, after IRBP-specific T cell injection, Faslpr mice did not release HMGB1 in the eye or develop EAU, but intravitreous injection of HMGB1 resulted in intraocular inflammation. Finally, tEAU in Wt mice was attenuated by local treatment with Met 12. Unlike HMGB1, Fas-induced IL-1 and IL-18 were not essential for tEAU induction. Conclusion Our results show that interaction of retinal cells with infiltrating uveitogenic T cells leads to rapid release of HMGB1 via the Fas/FasL inflammatory signaling pathway.
Collapse
Affiliation(s)
- Guomin Jiang
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, 301 E. Muhammad Ali Blvd., Louisville, KY, 40202, USA
| | - Yunsong Wang
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, 301 E. Muhammad Ali Blvd., Louisville, KY, 40202, USA.,Department of Ophthalmology, Tangshan Gongren Hospital, Tangshan, Hebei, 063000, China
| | - Juan Yun
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, 301 E. Muhammad Ali Blvd., Louisville, KY, 40202, USA.,Department of Pharmaceutical Sciences, Sullivan University College of Pharmacy, Louisville, KY, 40205, USA
| | - Amir Reza Hajrasouliha
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, 301 E. Muhammad Ali Blvd., Louisville, KY, 40202, USA
| | - Yuan Zhao
- Department of Pharmaceutical Sciences, Sullivan University College of Pharmacy, Louisville, KY, 40205, USA
| | - Deming Sun
- Doheny Eye Institute, Los Angeles, CA, 90033, USA
| | - Henry J Kaplan
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, 301 E. Muhammad Ali Blvd., Louisville, KY, 40202, USA
| | - Hui Shao
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, 301 E. Muhammad Ali Blvd., Louisville, KY, 40202, USA.
| |
Collapse
|
30
|
Incrocci R, Hussain S, Stone A, Bieging K, Alt LAC, Fay MJ, Swanson-Mungerson M. Epstein-Barr virus Latent Membrane Protein 2A (LMP2A)-mediated changes in Fas expression and Fas-dependent apoptosis: Role of Lyn/Syk activation. Cell Immunol 2015; 297:108-19. [PMID: 26255694 DOI: 10.1016/j.cellimm.2015.08.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 07/15/2015] [Accepted: 08/04/2015] [Indexed: 01/01/2023]
Abstract
Epstein-Barr virus Latent Membrane Protein 2A (LMP2A) is expressed in EBV-infected B cells in the germinal center, a site of significant apoptosis induced by engagement of Fas on activated B cells. Signals from the B cell receptor (BCR) protect germinal center B cells from Fas-mediated apoptosis, and since LMP2A is a BCR mimic, we hypothesized that LMP2A would also protect B cells from Fas-mediated apoptosis. Surprisingly, latently-infected human and murine B cell lines expressing LMP2A were more sensitive to Fas-mediated apoptosis, as determined by increases in Annexin-V staining, and cleavage of caspase-8, -3 and PARP. Additional studies show that LMP2A-expressing B cell lines demonstrate a Lyn- and Syk-dependent increase in sensitivity to Fas-mediated apoptosis, due to an LMP2A-dependent enhancement in Fas expression. These findings demonstrate the ability for LMP2A to directly increase a pro-apoptotic molecule and have implications for EBV latency as well as the treatment of EBV-associated malignancies.
Collapse
Affiliation(s)
- Ryan Incrocci
- Chicago College of Osteopathic Medicine, Department of Microbiology and Immunology, 555 31st Street, Downers Grove, IL 60515, USA
| | - Samira Hussain
- College of Health Sciences, Department of Biomedical Sciences, 555 31st Street, Downers Grove, IL 60515, USA
| | - Amanda Stone
- Chicago College of Osteopathic Medicine, Department of Microbiology and Immunology, 555 31st Street, Downers Grove, IL 60515, USA
| | - Kathryn Bieging
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lauren A C Alt
- College of Health Sciences, Department of Biomedical Sciences, 555 31st Street, Downers Grove, IL 60515, USA
| | - Michael J Fay
- College of Health Sciences, Department of Biomedical Sciences, 555 31st Street, Downers Grove, IL 60515, USA
| | - Michelle Swanson-Mungerson
- Chicago College of Osteopathic Medicine, Department of Microbiology and Immunology, 555 31st Street, Downers Grove, IL 60515, USA.
| |
Collapse
|
31
|
Wajant H. Principles and mechanisms of CD95 activation. Biol Chem 2015; 395:1401-16. [PMID: 25153377 DOI: 10.1515/hsz-2014-0212] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 08/06/2014] [Indexed: 11/15/2022]
Abstract
CD95 (Apo1/Fas) has been originally identified as the target of cell death-inducing antibodies. The recognition of CD95 as an apoptosis-triggering receptor represents one of the early milestones in the apoptosis field. Moreover, the research on CD95-induced cell death fostered various other discoveries of broad and general relevance in cell biology, for example, the identification of caspase 8 as the initiator caspase of the extrinsic apoptosis pathway. Activation of CD95-associated intracellular signaling pathways is not a simple consequence of ligand binding but is the fine-tuned result of a complex interplay of various molecular mechanisms that eventually determine the strength and quality of the CD95 response. There is growing evidence that different forms of CD95 stimulation trigger the assembly of CD95 signaling complexes of distinct composition. Moreover, the formation of signaling competent CD95 complexes is a multistep process and the subject of regulation by various cellular cues. This review addresses the relevance of the molecular nature of the CD95-stimulating agonist for the quality of the CD95 response and discusses the importance of modification, clustering, internalization, and lipid raft and actin association of CD95 for CD95 activity.
Collapse
|
32
|
Zhou Y, Li Y, Mu T. HMGB1 Neutralizing Antibody Attenuates Cardiac Injury and Apoptosis Induced by Hemorrhagic Shock/Resuscitation in Rats. Biol Pharm Bull 2015; 38:1150-60. [PMID: 26040987 DOI: 10.1248/bpb.b15-00026] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
High-mobility group box 1 (HMGB1) and its natural receptor, Toll-like receptor-4 (TLR4), are involved in various infectious or noninfectious diseases including hemorrhagic shock. HMGB1 neutralizing antibody (anti-HMGB1 monoclonal antibody (mAb)) treatment was shown to alleviate ischemia-reperfusion injury effectively. The aim of this study was to explore whether and by what mechanisms anti-HMGB1 mAb attenuates hemorrhagic shock and resuscitation (HS/R)-induced cardiac injury. Employing rat HS/R models, we found that anti-HMGB1 mAb treatment improved HS/R-induced cardiac function deterioration, attenuated cardiac enzyme elevation, improved ATP loss, and protected cardiac tissue. Anti-HMGB1 mAb also inhibited the production of inflammatory factors interleukin (IL)-1β, IL-6, and tumor necrosis factor-α (TNF-α). Moreover, anti-HMGB1 mAb reduced apoptotic responses by suppressing activated caspase-3 and reversing apoptotic gene expression of capase-3, Bax, and Bcl-2 in rat cardiac tissue. Moreover, anti-HMGB1 mAb decreased HS/R-induced HMGB1 and TLR4 expression elevation. We further confirmed that anti-HMGB1 mAb inhibited lipopolysaccharide-activated HGMB1 and TLR4 expression and decreased inflammatory factors IL-1β, IL-6, and TNF-α at the cellular level. It was concluded that anti-HMGB1 mAb treatment protects rats from cardiac injury induced by HS/R, and the beneficial effects may be related to its inhibitory effects on the HMGB1-TLR4 axis.
Collapse
Affiliation(s)
- Yu Zhou
- Department of Orthopedics, Beijing Chao-Yang Hospital, Capital Medical University
| | | | | |
Collapse
|
33
|
Guzman-Flores JM, Cortez-Espinosa N, Cortés-Garcia JD, Vargas-Morales JM, Cataño-Cañizalez YG, Rodríguez-Rivera JG, Portales-Perez DP. Expression of CD73 and A2A receptors in cells from subjects with obesity and type 2 diabetes mellitus. Immunobiology 2015; 220:976-84. [PMID: 25770019 DOI: 10.1016/j.imbio.2015.02.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 02/10/2015] [Accepted: 02/19/2015] [Indexed: 01/01/2023]
Abstract
Regulatory T cells have various mechanisms to suppress the inflammatory response, among these, the modulation of the microenvironment through adenosine and with the participation of CD39, CD73 and A2A. The aim of this study was to assess the expression of CD73 and A2A in immune cells and the effect of activation of A2A by an adenosine analogue on apoptosis in patients with obesity and type 2 diabetes mellitus (T2D). CD73 and A2A expression were analyzed by flow cytometry in lymphocyte subpopulations from patients with obesity (n = 22), T2D (n = 22), and healthy subjects (n = 20). Lymphocytes were treated with the selective A2A antagonist (ZM241385) or the selective A2A agonist (CGS21680), and apoptotic cells were detected by Annexin V. We found an increased expression of CD39 coupled to a decrease in CD73 in the patient groups with obesity and T2D compared to the control group in the different studied lymphocyte subpopulations. A2A expression was found to be increased in different subpopulations of lymphocytes from T2D patients. We also detected positive correlations between CD39+ cells and age and BMI. Meanwhile, CD73+ cells showed negative correlations with age, WHR, BMI, FPG, HbAc1, triglycerides and cholesterol. Moreover, an increase in the percentage of apoptotic cells from T2D patients with regard to the groups with obesity and control was observed. In addition, the CD8+ T cells of patients with T2D exhibited decreased apoptosis when treated with the A2A agonist. In conclusion, our data suggest a possible role for CD73 and A2A in inflammation observed in patients with T2D and obesity mediated via apoptosis.
Collapse
Affiliation(s)
- Juan M Guzman-Flores
- Laboratory of Immunology and Cellular and Molecular Biology, Faculty of Chemical Sciences, UASLP, San Luis Potosí, S.L.P., Mexico
| | - Nancy Cortez-Espinosa
- Laboratory of Immunology and Cellular and Molecular Biology, Faculty of Chemical Sciences, UASLP, San Luis Potosí, S.L.P., Mexico
| | - Juan D Cortés-Garcia
- Laboratory of Immunology and Cellular and Molecular Biology, Faculty of Chemical Sciences, UASLP, San Luis Potosí, S.L.P., Mexico
| | - Juan M Vargas-Morales
- Laboratory of Clinical Analysis "Dr. Pedro Medina de los Santos", Faculty of Chemical Sciences, UASLP, San Luis Potosí, S.L.P., Mexico
| | - Yolanda G Cataño-Cañizalez
- Laboratory of Immunology and Cellular and Molecular Biology, Faculty of Chemical Sciences, UASLP, San Luis Potosí, S.L.P., Mexico
| | - Jaime G Rodríguez-Rivera
- Department of Internal Medicine and Endocrinology, Hospital Central Dr Ignacio MoronesPrieto, San Luis Potosí, S.L.P., Mexico
| | - Diana P Portales-Perez
- Laboratory of Immunology and Cellular and Molecular Biology, Faculty of Chemical Sciences, UASLP, San Luis Potosí, S.L.P., Mexico.
| |
Collapse
|
34
|
NF-κB regulates caspase-4 expression and sensitizes neuroblastoma cells to Fas-induced apoptosis. PLoS One 2015; 10:e0117953. [PMID: 25695505 PMCID: PMC4335045 DOI: 10.1371/journal.pone.0117953] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 01/06/2015] [Indexed: 12/01/2022] Open
Abstract
Found in neurons and neuroblastoma cells, Fas-induced apoptosis and accompanied activation of NF-κB signaling were thought to be associated with neurodegenerative diseases. However, the detailed functions of NF-κB activation in Fas killing and the effect of NF-κB activation on its downstream events remain unclear. Here, we demonstrated that agonistic Fas antibody induces cell death in a dose-dependent way and NF-κB signaling is activated as well, in neuroblastoma cells SH-EP1. Unexpectedly, NF-κB activation was shown to be pro-apoptotic, as suggested by the reduction of Fas-induced cell death with either a dominant negative form of IκBα (DN-IκBα) or an IκB kinase-specific inhibitor. To our interest, when analyzing downstream events of NF-κB signaling, we found that DN-IκBα only suppressed the expression of caspase-4, but not other caspases. Vice versa, enhancement of NF-κB activity by p65 (RelA) overexpression increased the expression of caspase-4 at both mRNA and protein levels. More directly, results from dual luciferase reporter assay demonstrated the regulation of caspase-4 promoter activity by NF-κB. When caspase-4 activity was blocked by its dominant negative (DN) form, Fas-induced cell death was substantially reduced. Consistently, the cleavage of PARP and caspase-3 induced by Fas was also reduced. In contrast, the cleavage of caspase-8 remained unaffected in caspase-4 DN cells, although caspase-8 inhibitor could rescue Fas-induced cell death. Collectively, these data suggest that caspase-4 activity is required for Fas-induced cell apoptosis and caspase-4 may act upstream of PARP and caspase-3 and downstream of caspase-8. Overall, we demonstrate that NF-κB can mediate Fas-induced apoptosis through caspase-4 protease, indicating that caspase-4 is a new mediator of NF-κB pro-apoptotic pathway in neuroblastoma cells.
Collapse
|
35
|
Yu AK, Song L, Murray KD, van der List D, Sun C, Shen Y, Xia Z, Cortopassi GA. Mitochondrial complex I deficiency leads to inflammation and retinal ganglion cell death in the Ndufs4 mouse. Hum Mol Genet 2015; 24:2848-60. [PMID: 25652399 DOI: 10.1093/hmg/ddv045] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 02/02/2015] [Indexed: 11/13/2022] Open
Abstract
Mitochondrial complex I (NADH dehydrogenase) is a major contributor to neuronal energetics, and mutations in complex I lead to vision loss. Functional, neuroanatomical and transcriptional consequences of complex I deficiency were investigated in retinas of the Ndufs4 knockout mouse. Whole-eye ERGs and multielectrode arrays confirmed a major retinal ganglion cell functional loss at P32, and retinal ganglion cell loss at P42. RNAseq demonstrated a mild and then sharp increase in innate immune and inflammatory retinal transcripts at P22 and P33, respectively, which were confirmed with QRT-PCR. Intraperitoneal injection of the inflammogen lipopolysaccharide further reduced retinal ganglion cell function in Ndufs4 KO, supporting the connection between inflammatory activation and functional loss. Complex I deficiency in the retina clearly caused innate immune and inflammatory markers to increase coincident with loss of vision, and RGC functional loss. How complex I incites inflammation and functional loss is not clear, but could be the result of misfolded complex I generating a 'non-self' response, and induction of innate immune response transcripts was observed before functional loss at P22, including β-2 microglobulin and Cx3cr1, and during vision loss at P31 (B2m, Tlr 2, 3, 4, C1qa, Cx3cr1 and Fas). These data support the hypothesis that mitochondrial complex I dysfunction in the retina triggers an innate immune and inflammatory response that results in loss of retinal ganglion cell function and death, as in Leber's hereditary Optic Neuropathy and suggests novel therapeutic routes to counter mitochondrial defects that contribute to vision loss.
Collapse
Affiliation(s)
| | | | | | | | - Chao Sun
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA 95616, USA and
| | - Yan Shen
- Veterinary Medicine, Molecular Biosciences
| | - Zhengui Xia
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | | |
Collapse
|
36
|
XIAP-targeting drugs re-sensitize PIK3CA-mutated colorectal cancer cells for death receptor-induced apoptosis. Cell Death Dis 2014; 5:e1570. [PMID: 25501831 PMCID: PMC4649844 DOI: 10.1038/cddis.2014.534] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 10/17/2014] [Accepted: 11/05/2014] [Indexed: 02/07/2023]
Abstract
Mutations in the oncogenic PIK3CA gene are found in 10–20% of colorectal cancers (CRCs) and are associated with poor prognosis. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and agonistic TRAIL death receptor antibodies emerged as promising anti-neoplastic therapeutics, but to date failed to prove their capability in the clinical setting as especially primary tumors exhibit high rates of TRAIL resistance. In our study, we investigated the molecular mechanisms underlying TRAIL resistance in CRC cells with a mutant PIK3CA (PIK3CA-mut) gene. We show that inhibition of the constitutively active phosphatidylinositol-3 kinase (PI3K)/Akt signaling pathway only partially overcame TRAIL resistance in PIK3CA-mut-protected HCT116 cells, although synergistic effects of TRAIL plus PI3K, Akt or cyclin-dependent kinase (CDK) inhibitors could be noted. In sharp contrast, TRAIL triggered full-blown cell death induction in HCT116 PIK3CA-mut cells treated with proteasome inhibitors such as bortezomib and MG132. At the molecular level, resistance of HCT116 PIK3CA-mut cells against TRAIL was reflected by impaired caspase-3 activation and we provide evidence for a crucial involvement of the E3-ligase X-linked inhibitor of apoptosis protein (XIAP) therein. Drugs interfering with the activity and/or the expression of XIAP, such as the second mitochondria-derived activator of caspase mimetic BV6 and mithramycin-A, completely restored TRAIL sensitivity in PIK3CA-mut-protected HCT116 cells independent of a functional mitochondrial cell death pathway. Importantly, proteasome inhibitors and XIAP-targeting agents also sensitized other CRC cell lines with mutated PIK3CA for TRAIL-induced cell death. Together, our data suggest that proteasome- or XIAP-targeting drugs offer a novel therapeutic approach to overcome TRAIL resistance in PIK3CA-mutated CRC.
Collapse
|
37
|
Regulators of carcinogenesis: emerging roles beyond their primary functions. Cancer Lett 2014; 357:75-82. [PMID: 25448403 DOI: 10.1016/j.canlet.2014.11.048] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Revised: 11/23/2014] [Accepted: 11/24/2014] [Indexed: 12/20/2022]
Abstract
Cancers are characterized by aberrant cell signaling that results in accelerated proliferation, suppressed cell death, and reprogrammed metabolism to provide sufficient energy and intermediate metabolites for macromolecular biosynthesis. Here, we summarize the emerging "unconventional" roles of these regulators based on their newly identified interaction partners, different subcellular localizations, and/or structural variants. For example, the epidermal growth factor receptor (EGFR) regulates DNA synthesis, microRNA maturation and drug resistance by interacting with previously undescribed partners; cyclins and cyclin-dependent kinases (CDKs) crosstalk with multiple canonical pathways by phosphorylating novel substrates or by functioning as transcriptional factors; apoptosis executioners play extensive roles in necroptosis, autophagy, and in the self-renewal of stem cells; and various metabolic enzymes and their mutants control carcinogenesis independently of their enzymatic activity. These recent findings will supplement the systemic functional annotation of cancer regulators and provide new rationales for potential molecular targeted cancer treatments.
Collapse
|
38
|
Fernandes P, O’Donnell C, Lyons C, Keane J, Regan T, O’Brien S, Fallon P, Brint E, Houston A. Intestinal Expression of Fas and Fas Ligand Is Upregulated by Bacterial Signaling through TLR4 and TLR5, with Activation of Fas Modulating Intestinal TLR-Mediated Inflammation. THE JOURNAL OF IMMUNOLOGY 2014; 193:6103-13. [DOI: 10.4049/jimmunol.1303083] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
39
|
Song S, Chen D, Ma T, Luo Y, Yang Z, Wang D, Fan X, Qin Q, Ni B, Guo X, Xian Z, Lan P, Cao X, Li M, Wang J, Wang L. Molecular mechanism of acute radiation enteritis revealed using proteomics and biological signaling network analysis in rats. Dig Dis Sci 2014; 59:2704-13. [PMID: 24927798 DOI: 10.1007/s10620-014-3224-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 05/21/2014] [Indexed: 12/09/2022]
Abstract
BACKGROUND AND AIMS Radiation enteritis (RE) has emerged as a significant complication that can progress to severe gastrointestinal disease and the mechanisms underlying its genesis remain poorly understood. The aim of this study was to identify temporal changes in protein expression potentially associated with acute inflammation and to elucidate the mechanism underlying radiation enteritis genesis. METHODS Male Sprague-Dawley rats were irradiated in the abdomen with a single dose of 10 Gy to establish an in vivo model of acute radiation enteritis. Two-dimensional fluorescence difference gel electrophoresis, matrix-assisted laser desorption/ionization time-of-flight spectrometer (MALDI-TOF) tandem mass spectrometry, and peptide mass fingerprinting were used to determine differentially expressed proteins between normal and inflamed intestinal mucosa. Additionally, differentially expressed proteins were evaluated by KO Based Annotation System to find the biological functions associated with acute radiation enteritis. RESULTS Intensity changes of 86 spots were detected with statistical significance (ratio ≥ 1.5 or ≤ 1.5, P < 0.05). Sixty one of the 86 spots were identified by MALDI-TOF/TOF tandem mass spectrometry. These radiation-induced proteins with biological functions showed that the FAS pathway and glycolysis signaling pathways were significantly altered using the KOBAS tool. CONCLUSIONS Our results reveal an underlying mechanism of radiation-induced acute enteritis, which may help clarify the pathogenesis of RE and point to potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- Shunxin Song
- Gastrointestinal Institute of Sun Yat-Sen University, The Sixth Affiliated Hospital of Sun Yat-Sen University, 26 Yuancunerheng Road, Guangzhou, 510655, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Campisi L, Cummings RJ, Blander JM. Death-defining immune responses after apoptosis. Am J Transplant 2014; 14:1488-98. [PMID: 24903539 PMCID: PMC4115279 DOI: 10.1111/ajt.12736] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 03/04/2014] [Accepted: 03/10/2014] [Indexed: 01/25/2023]
Abstract
Apoptosis is a programmed form of cell death whereby characteristic internal cellular dismantling is accompanied by the preservation of plasma membrane integrity. Maintaining this order during apoptosis prevents the release of cellular contents and ensures a noninflammatory death. Here, we consider examples of apoptosis in different contexts and discuss how the same form of cell death could have different immunological consequences. Multiple parameters such as cell death as a result of microbial infection, the nature of the inflammatory microenvironment, the type of responding phagocytic cells and the genetic background of the host organism all differentially influence the immunological consequences of apoptosis.
Collapse
|
41
|
Zhi F, Zhou G, Shao N, Xia X, Shi Y, Wang Q, Zhang Y, Wang R, Xue L, Wang S, Wu S, Peng Y, Yang Y. miR-106a-5p inhibits the proliferation and migration of astrocytoma cells and promotes apoptosis by targeting FASTK. PLoS One 2013; 8:e72390. [PMID: 24013584 PMCID: PMC3754986 DOI: 10.1371/journal.pone.0072390] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 07/09/2013] [Indexed: 01/11/2023] Open
Abstract
Astrocytomas are common malignant intracranial tumors that comprise the majority of adult primary central nervous system tumors. MicroRNAs (miRNAs) are small, non-coding RNAs (20–24 nucleotides) that post-transcriptionally modulate gene expression by negatively regulating the stability or translational efficiency of their target mRNAs. In our previous studies, we found that the downregulation of miR-106a-5p in astrocytomas is associated with poor prognosis. However, its specific gene target(s) and underlying functional mechanism(s) in astrocytomas remain unclear. In this study, we used mRNA microarray experiments to measure global mRNA expression in the presence of increased or decreased miR-106a-5p levels. We then performed bioinformatics analysis based on multiple target prediction algorithms to obtain candidate target genes that were further validated by computational predictions, western blot analysis, quantitative real-time PCR, and the luciferase reporter assay. Fas-activated serine/threonine kinase (FASTK) was identified as a direct target of miR-106a-5p. In human astrocytomas, miR-106a-5p is downregulated and negatively associated with clinical staging, whereas FASTK is upregulated and positively associated with advanced clinical stages, at both the protein and mRNA levels. Furthermore, Kaplan-Meier analysis revealed that the reduced expression of miR-106a-5p or the increased expression of FASTK is significantly associated with poor survival outcome. These results further supported the finding that FASTK is a direct target gene of miR-106a-5p. Next, we explored the function of miR-106a-5p and FASTK during astrocytoma progression. Through gain-of-function and loss-of-function studies, we demonstrated that miR-106a-5p can significantly inhibit cell proliferation and migration and can promote cell apoptosis in vitro. The knockdown of FASTK induced similar effects on astrocytoma cells as those induced by the overexpression of miR-106a-5p. These observations suggest that miR-106a-5p functions as a tumor suppressor during the development of astrocytomas by targeting FASTK.
Collapse
Affiliation(s)
- Feng Zhi
- Modern Medical Research Center, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Guangxin Zhou
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Naiyuan Shao
- Department of Neurosurgery, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Xiwei Xia
- Department of Neurosurgery, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Yimin Shi
- Department of Neurosurgery, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Qiang Wang
- Department of Neurosurgery, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Yi Zhang
- Department of Neurosurgery, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Rong Wang
- Modern Medical Research Center, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Lian Xue
- Modern Medical Research Center, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Suinuan Wang
- Department of Neurosurgery, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Sujia Wu
- Department of Orthopedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
- * E-mail: (YY); (YP); (SW)
| | - Ya Peng
- Department of Neurosurgery, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- * E-mail: (YY); (YP); (SW)
| | - Yilin Yang
- Modern Medical Research Center, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- * E-mail: (YY); (YP); (SW)
| |
Collapse
|