51
|
Khandibharad S, Nimsarkar P, Singh S. Mechanobiology of immune cells: Messengers, receivers and followers in leishmaniasis aiding synthetic devices. CURRENT RESEARCH IN IMMUNOLOGY 2022; 3:186-198. [PMID: 36051499 PMCID: PMC9424266 DOI: 10.1016/j.crimmu.2022.08.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/04/2022] [Accepted: 08/10/2022] [Indexed: 11/03/2022] Open
Abstract
Cytokines are influential molecules which can direct cells behavior. In this review, cytokines are referred as messengers, immune cells which respond to cytokine stimulus are referred as receivers and the immune cells which gets modulated due to their plasticity induced by infectious pathogen leishmania, are referred as followers. The advantage of plasticity of cells is taken by the parasite to switch them from parasite eliminating form to parasite survival favoring form through a process called as reciprocity which is undergone by cytokines, wherein pro-inflammatory to anti-inflammatory switch occur rendering immune cell population to switch their phenotype. Detailed study of this switch can help in identification of important targets which can help in restoring the phenotype to parasite eliminating form and this can be done through synthetic circuit, finding its wider applicability in therapeutics. Cytokines as messengers for governing reciprocity in infection. Leishmania induces reciprocity modulating the immune cells plasticity. Reciprocity of cytokines identifies important target for therapeutics. Therapeutic targets aiding the design of synthetic devices to combat infection.
Collapse
|
52
|
B Cell Involvement in the Pathogenesis of Ankylosing Spondylitis. Int J Mol Sci 2021; 22:ijms222413325. [PMID: 34948121 PMCID: PMC8703482 DOI: 10.3390/ijms222413325] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 12/25/2022] Open
Abstract
Extensive research into ankylosing spondylitis (AS) has suggested the major role of genetics, immune reactions, and the joint-gut axis in its etiology, although an ultimate consensus does not yet exist. The available evidence indicates that both autoinflammation and T-cell-mediated autoimmune processes are actively involved in the disease process of AS. So far, B cells have received relatively little attention in AS pathogenesis; this is largely due to a lack of conventional disease-defining autoantibodies. However, against prevailing dogma, there is a growing body of evidence suggestive of B cell involvement. This is illustrated by disturbances in circulating B cell populations and the formation of auto-reactive and non-autoreactive antibodies, along with B cell infiltrates within the axial skeleton of AS patients. Furthermore, the depletion of B cells, using rituximab, displayed beneficial results in a subgroup of patients with AS. This review provides an overview of our current knowledge of B cells in AS, and discusses their potential role in its pathogenesis. An overarching picture portrays increased B cell activation in AS, although it is unclear whether B cells directly affect pathogenesis, or are merely bystanders in the disease process.
Collapse
|
53
|
Chipurupalli S, Samavedam U, Robinson N. Crosstalk Between ER Stress, Autophagy and Inflammation. Front Med (Lausanne) 2021; 8:758311. [PMID: 34805224 PMCID: PMC8602556 DOI: 10.3389/fmed.2021.758311] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 10/14/2021] [Indexed: 12/23/2022] Open
Abstract
The endoplasmic reticulum (ER) is not only responsible for protein synthesis and folding but also plays a critical role in sensing cellular stress and maintaining cellular homeostasis. Upon sensing the accumulation of unfolded proteins due to perturbation in protein synthesis or folding, specific intracellular signaling pathways are activated, which are collectively termed as unfolded protein response (UPR). UPR expands the capacity of the protein folding machinery, decreases protein synthesis and enhances ER-associated protein degradation (ERAD) which degrades misfolded proteins through the proteasomes. More recent evidences suggest that UPR also amplifies cytokines-mediated inflammatory responses leading to pathogenesis of inflammatory diseases. UPR signaling also activates autophagy; a lysosome-dependent degradative pathwaythat has an extended capacity to degrade misfolded proteins and damaged ER. Thus, activation of autophagy limits inflammatory response and provides cyto-protection by attenuating ER-stress. Here we review the mechanisms that couple UPR, autophagy and cytokine-induced inflammation that can facilitate the development of novel therapeutic strategies to mitigate cellular stress and inflammation associated with various pathologies.
Collapse
Affiliation(s)
- Sandhya Chipurupalli
- Cellular-Stress and Immune Response Laboratory, Center for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia
| | - Unni Samavedam
- College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Nirmal Robinson
- Cellular-Stress and Immune Response Laboratory, Center for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia
| |
Collapse
|
54
|
Trouvé P, Férec C, Génin E. The Interplay between the Unfolded Protein Response, Inflammation and Infection in Cystic Fibrosis. Cells 2021; 10:2980. [PMID: 34831204 PMCID: PMC8616505 DOI: 10.3390/cells10112980] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 10/25/2021] [Accepted: 10/29/2021] [Indexed: 12/15/2022] Open
Abstract
In cystic fibrosis (CF), p.Phe508del is the most frequent mutation in the Cystic Fibrosis Transmembrane conductance Regulator (CFTR) gene. The p.Phe508del-CFTR protein is retained in the ER and rapidly degraded. This retention likely triggers an atypical Unfolded Protein Response (UPR) involving ATF6, which reduces the expression of p.Phe508del-CFTR. There are still some debates on the role of the UPR in CF: could it be triggered by the accumulation of misfolded CFTR proteins in the endoplasmic reticulum as was proposed for the most common CFTR mutation p.Phe508del? Or, is it the consequence of inflammation and infection that occur in the disease? In this review, we summarize recent findings on UPR in CF and show how infection, inflammation and UPR act together in CF. We propose to rethink their respective role in CF and to consider them as a whole.
Collapse
Affiliation(s)
- Pascal Trouvé
- Inserm, Univ Brest, EFS, UMR 1078, GGB, F-29200 Brest, France; (C.F.); (E.G.)
| | | | | |
Collapse
|
55
|
Sayce AC, Martinez FO, Tyrrell BE, Perera N, Hill ML, Dwek RA, Miller JL, Zitzmann N. Pathogen-induced inflammation is attenuated by the iminosugar MON-DNJ via modulation of the unfolded protein response. Immunology 2021; 164:587-601. [PMID: 34287854 PMCID: PMC8517592 DOI: 10.1111/imm.13393] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/17/2021] [Accepted: 06/28/2021] [Indexed: 12/27/2022] Open
Abstract
Sepsis is a life-threatening condition involving a dysregulated immune response to infectious agents that cause injury to host tissues and organs. Current treatments are limited to early administration of antibiotics and supportive care. While appealing, the strategy of targeted inhibition of individual molecules in the inflammatory cascade has not proved beneficial. Non-targeted, systemic immunosuppression with steroids has shown limited efficacy and raises concern for secondary infection. Iminosugars are a class of small molecule glycomimetics with distinct inhibition profiles for glycan processing enzymes based on stereochemistry. Inhibition of host endoplasmic reticulum resident glycoprotein processing enzymes has demonstrated efficacy as a broad-spectrum antiviral strategy, but limited consideration has been given to the effects on host glycoprotein production and consequent disruption of signalling cascades. This work demonstrates that iminosugars inhibit dengue virus, bacterial lipopolysaccharide and fungal antigen-stimulated cytokine responses in human macrophages. In spite of decreased inflammatory mediator production, viral replication is suppressed in the presence of iminosugar. Transcriptome analysis reveals the key interaction of pathogen-induced endoplasmic reticulum stress, the resulting unfolded protein response and inflammation. Our work shows that iminosugars modulate these interactions. Based on these findings, we propose a new therapeutic role for iminosugars as treatment for sepsis-related inflammatory disorders associated with excess cytokine secretion.
Collapse
Affiliation(s)
- Andrew C. Sayce
- Oxford Glycobiology InstituteDepartment of BiochemistryUniversity of OxfordOxfordUK
- Vanderbilt University School of MedicineVanderbilt UniversityNashvilleTennesseeUSA
| | | | - Beatrice E. Tyrrell
- Oxford Glycobiology InstituteDepartment of BiochemistryUniversity of OxfordOxfordUK
| | - Nilanka Perera
- Oxford Glycobiology InstituteDepartment of BiochemistryUniversity of OxfordOxfordUK
- Faculty of Medical SciencesUniversity of Sri JayewardenepuraGangodawilaNugegodaSri Lanka
| | - Michelle L. Hill
- Oxford Glycobiology InstituteDepartment of BiochemistryUniversity of OxfordOxfordUK
| | - Raymond A. Dwek
- Oxford Glycobiology InstituteDepartment of BiochemistryUniversity of OxfordOxfordUK
| | - Joanna L. Miller
- Oxford Glycobiology InstituteDepartment of BiochemistryUniversity of OxfordOxfordUK
| | - Nicole Zitzmann
- Oxford Glycobiology InstituteDepartment of BiochemistryUniversity of OxfordOxfordUK
| |
Collapse
|
56
|
Ostaszewski M, Niarakis A, Mazein A, Kuperstein I, Phair R, Orta‐Resendiz A, Singh V, Aghamiri SS, Acencio ML, Glaab E, Ruepp A, Fobo G, Montrone C, Brauner B, Frishman G, Monraz Gómez LC, Somers J, Hoch M, Kumar Gupta S, Scheel J, Borlinghaus H, Czauderna T, Schreiber F, Montagud A, Ponce de Leon M, Funahashi A, Hiki Y, Hiroi N, Yamada TG, Dräger A, Renz A, Naveez M, Bocskei Z, Messina F, Börnigen D, Fergusson L, Conti M, Rameil M, Nakonecnij V, Vanhoefer J, Schmiester L, Wang M, Ackerman EE, Shoemaker JE, Zucker J, Oxford K, Teuton J, Kocakaya E, Summak GY, Hanspers K, Kutmon M, Coort S, Eijssen L, Ehrhart F, Rex DAB, Slenter D, Martens M, Pham N, Haw R, Jassal B, Matthews L, Orlic‐Milacic M, Senff Ribeiro A, Rothfels K, Shamovsky V, Stephan R, Sevilla C, Varusai T, Ravel J, Fraser R, Ortseifen V, Marchesi S, Gawron P, Smula E, Heirendt L, Satagopam V, Wu G, Riutta A, Golebiewski M, Owen S, Goble C, Hu X, Overall RW, Maier D, Bauch A, Gyori BM, Bachman JA, Vega C, Grouès V, Vazquez M, Porras P, Licata L, Iannuccelli M, Sacco F, Nesterova A, Yuryev A, de Waard A, Turei D, Luna A, Babur O, Soliman S, Valdeolivas A, Esteban‐Medina M, Peña‐Chilet M, Rian K, Helikar T, Puniya BL, Modos D, Treveil A, Olbei M, De Meulder B, Ballereau S, Dugourd A, Naldi A, Noël V, Calzone L, Sander C, Demir E, Korcsmaros T, Freeman TC, Augé F, Beckmann JS, Hasenauer J, Wolkenhauer O, Wilighagen EL, Pico AR, Evelo CT, Gillespie ME, Stein LD, Hermjakob H, D'Eustachio P, Saez‐Rodriguez J, Dopazo J, Valencia A, Kitano H, Barillot E, Auffray C, Balling R, Schneider R. COVID19 Disease Map, a computational knowledge repository of virus-host interaction mechanisms. Mol Syst Biol 2021; 17:e10387. [PMID: 34664389 PMCID: PMC8524328 DOI: 10.15252/msb.202110387] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 12/13/2022] Open
Abstract
We need to effectively combine the knowledge from surging literature with complex datasets to propose mechanistic models of SARS-CoV-2 infection, improving data interpretation and predicting key targets of intervention. Here, we describe a large-scale community effort to build an open access, interoperable and computable repository of COVID-19 molecular mechanisms. The COVID-19 Disease Map (C19DMap) is a graphical, interactive representation of disease-relevant molecular mechanisms linking many knowledge sources. Notably, it is a computational resource for graph-based analyses and disease modelling. To this end, we established a framework of tools, platforms and guidelines necessary for a multifaceted community of biocurators, domain experts, bioinformaticians and computational biologists. The diagrams of the C19DMap, curated from the literature, are integrated with relevant interaction and text mining databases. We demonstrate the application of network analysis and modelling approaches by concrete examples to highlight new testable hypotheses. This framework helps to find signatures of SARS-CoV-2 predisposition, treatment response or prioritisation of drug candidates. Such an approach may help deal with new waves of COVID-19 or similar pandemics in the long-term perspective.
Collapse
Affiliation(s)
- Marek Ostaszewski
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Anna Niarakis
- Université Paris‐SaclayLaboratoire Européen de Recherche pour la Polyarthrite rhumatoïde ‐ GenhotelUniv EvryEvryFrance
- Lifeware GroupInria Saclay‐Ile de FrancePalaiseauFrance
| | - Alexander Mazein
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Inna Kuperstein
- Institut CuriePSL Research UniversityParisFrance
- INSERMParisFrance
- MINES ParisTechPSL Research UniversityParisFrance
| | - Robert Phair
- Integrative Bioinformatics, Inc.Mountain ViewCAUSA
| | - Aurelio Orta‐Resendiz
- Institut PasteurUniversité de Paris, Unité HIVInflammation et PersistanceParisFrance
- Bio Sorbonne Paris CitéUniversité de ParisParisFrance
| | - Vidisha Singh
- Université Paris‐SaclayLaboratoire Européen de Recherche pour la Polyarthrite rhumatoïde ‐ GenhotelUniv EvryEvryFrance
| | - Sara Sadat Aghamiri
- Inserm‐ Institut national de la santé et de la recherche médicaleParisFrance
| | - Marcio Luis Acencio
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Enrico Glaab
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Andreas Ruepp
- Institute of Experimental Genetics (IEG)Helmholtz Zentrum München‐German Research Center for Environmental Health (GmbH)NeuherbergGermany
| | - Gisela Fobo
- Institute of Experimental Genetics (IEG)Helmholtz Zentrum München‐German Research Center for Environmental Health (GmbH)NeuherbergGermany
| | - Corinna Montrone
- Institute of Experimental Genetics (IEG)Helmholtz Zentrum München‐German Research Center for Environmental Health (GmbH)NeuherbergGermany
| | - Barbara Brauner
- Institute of Experimental Genetics (IEG)Helmholtz Zentrum München‐German Research Center for Environmental Health (GmbH)NeuherbergGermany
| | - Goar Frishman
- Institute of Experimental Genetics (IEG)Helmholtz Zentrum München‐German Research Center for Environmental Health (GmbH)NeuherbergGermany
| | - Luis Cristóbal Monraz Gómez
- Institut CuriePSL Research UniversityParisFrance
- INSERMParisFrance
- MINES ParisTechPSL Research UniversityParisFrance
| | - Julia Somers
- Department of Molecular and Medical GeneticsOregon Health & Sciences UniversityPortlandORUSA
| | - Matti Hoch
- Department of Systems Biology and BioinformaticsUniversity of RostockRostockGermany
| | | | - Julia Scheel
- Department of Systems Biology and BioinformaticsUniversity of RostockRostockGermany
| | - Hanna Borlinghaus
- Department of Computer and Information ScienceUniversity of KonstanzKonstanzGermany
| | - Tobias Czauderna
- Faculty of Information TechnologyDepartment of Human‐Centred ComputingMonash UniversityClaytonVic.Australia
| | - Falk Schreiber
- Department of Computer and Information ScienceUniversity of KonstanzKonstanzGermany
- Faculty of Information TechnologyDepartment of Human‐Centred ComputingMonash UniversityClaytonVic.Australia
| | | | | | - Akira Funahashi
- Department of Biosciences and InformaticsKeio UniversityYokohamaJapan
| | - Yusuke Hiki
- Department of Biosciences and InformaticsKeio UniversityYokohamaJapan
| | - Noriko Hiroi
- Graduate School of Media and GovernanceResearch Institute at SFCKeio UniversityKanagawaJapan
| | - Takahiro G Yamada
- Department of Biosciences and InformaticsKeio UniversityYokohamaJapan
| | - Andreas Dräger
- Computational Systems Biology of Infections and Antimicrobial‐Resistant PathogensInstitute for Bioinformatics and Medical Informatics (IBMI)University of TübingenTübingenGermany
- Department of Computer ScienceUniversity of TübingenTübingenGermany
- German Center for Infection Research (DZIF), partner siteTübingenGermany
| | - Alina Renz
- Computational Systems Biology of Infections and Antimicrobial‐Resistant PathogensInstitute for Bioinformatics and Medical Informatics (IBMI)University of TübingenTübingenGermany
- Department of Computer ScienceUniversity of TübingenTübingenGermany
| | - Muhammad Naveez
- Department of Systems Biology and BioinformaticsUniversity of RostockRostockGermany
- Institute of Applied Computer SystemsRiga Technical UniversityRigaLatvia
| | - Zsolt Bocskei
- Sanofi R&DTranslational SciencesChilly‐MazarinFrance
| | - Francesco Messina
- Dipartimento di Epidemiologia Ricerca Pre‐Clinica e Diagnostica AvanzataNational Institute for Infectious Diseases 'Lazzaro Spallanzani' I.R.C.C.S.RomeItaly
- COVID‐19 INMI Network Medicine for IDs Study GroupNational Institute for Infectious Diseases 'Lazzaro Spallanzani' I.R.C.C.SRomeItaly
| | - Daniela Börnigen
- Bioinformatics Core FacilityUniversitätsklinikum Hamburg‐EppendorfHamburgGermany
| | - Liam Fergusson
- Royal (Dick) School of Veterinary MedicineThe University of EdinburghEdinburghUK
| | - Marta Conti
- Faculty of Mathematics and Natural SciencesUniversity of BonnBonnGermany
| | - Marius Rameil
- Faculty of Mathematics and Natural SciencesUniversity of BonnBonnGermany
| | - Vanessa Nakonecnij
- Faculty of Mathematics and Natural SciencesUniversity of BonnBonnGermany
| | - Jakob Vanhoefer
- Faculty of Mathematics and Natural SciencesUniversity of BonnBonnGermany
| | - Leonard Schmiester
- Faculty of Mathematics and Natural SciencesUniversity of BonnBonnGermany
- Center for MathematicsChair of Mathematical Modeling of Biological SystemsTechnische Universität MünchenGarchingGermany
| | - Muying Wang
- Department of Chemical and Petroleum EngineeringUniversity of PittsburghPittsburghPAUSA
| | - Emily E Ackerman
- Department of Chemical and Petroleum EngineeringUniversity of PittsburghPittsburghPAUSA
| | - Jason E Shoemaker
- Department of Chemical and Petroleum EngineeringUniversity of PittsburghPittsburghPAUSA
- Department of Computational and Systems BiologyUniversity of PittsburghPittsburghPAUSA
| | | | | | | | | | | | - Kristina Hanspers
- Institute of Data Science and BiotechnologyGladstone InstitutesSan FranciscoCAUSA
| | - Martina Kutmon
- Department of Bioinformatics ‐ BiGCaTNUTRIMMaastricht UniversityMaastrichtThe Netherlands
- Maastricht Centre for Systems Biology (MaCSBio)Maastricht UniversityMaastrichtThe Netherlands
| | - Susan Coort
- Department of Bioinformatics ‐ BiGCaTNUTRIMMaastricht UniversityMaastrichtThe Netherlands
| | - Lars Eijssen
- Department of Bioinformatics ‐ BiGCaTNUTRIMMaastricht UniversityMaastrichtThe Netherlands
- Maastricht University Medical CentreMaastrichtThe Netherlands
| | - Friederike Ehrhart
- Department of Bioinformatics ‐ BiGCaTNUTRIMMaastricht UniversityMaastrichtThe Netherlands
- Maastricht University Medical CentreMaastrichtThe Netherlands
| | | | - Denise Slenter
- Department of Bioinformatics ‐ BiGCaTNUTRIMMaastricht UniversityMaastrichtThe Netherlands
| | - Marvin Martens
- Department of Bioinformatics ‐ BiGCaTNUTRIMMaastricht UniversityMaastrichtThe Netherlands
| | - Nhung Pham
- Department of Bioinformatics ‐ BiGCaTNUTRIMMaastricht UniversityMaastrichtThe Netherlands
| | - Robin Haw
- MaRS CentreOntario Institute for Cancer ResearchTorontoONCanada
| | - Bijay Jassal
- MaRS CentreOntario Institute for Cancer ResearchTorontoONCanada
| | | | | | - Andrea Senff Ribeiro
- MaRS CentreOntario Institute for Cancer ResearchTorontoONCanada
- Universidade Federal do ParanáCuritibaBrasil
| | - Karen Rothfels
- MaRS CentreOntario Institute for Cancer ResearchTorontoONCanada
| | | | - Ralf Stephan
- MaRS CentreOntario Institute for Cancer ResearchTorontoONCanada
| | - Cristoffer Sevilla
- European Bioinformatics Institute (EMBL‐EBI)European Molecular Biology LaboratoryHinxton, CambridgeshireUK
| | - Thawfeek Varusai
- European Bioinformatics Institute (EMBL‐EBI)European Molecular Biology LaboratoryHinxton, CambridgeshireUK
| | - Jean‐Marie Ravel
- INSERM UMR_S 1256Nutrition, Genetics, and Environmental Risk Exposure (NGERE)Faculty of Medicine of NancyUniversity of LorraineNancyFrance
- Laboratoire de génétique médicaleCHRU NancyNancyFrance
| | - Rupsha Fraser
- Queen's Medical Research InstituteThe University of EdinburghEdinburghUK
| | - Vera Ortseifen
- Senior Research Group in Genome Research of Industrial MicroorganismsCenter for BiotechnologyBielefeld UniversityBielefeldGermany
| | - Silvia Marchesi
- Department of Surgical ScienceUppsala UniversityUppsalaSweden
| | - Piotr Gawron
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
- Institute of Computing SciencePoznan University of TechnologyPoznanPoland
| | - Ewa Smula
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Laurent Heirendt
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Venkata Satagopam
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Guanming Wu
- Department of Medical Informatics and Clinical EpidemiologyOregon Health & Science UniversityPortlandORUSA
| | - Anders Riutta
- Institute of Data Science and BiotechnologyGladstone InstitutesSan FranciscoCAUSA
| | | | - Stuart Owen
- Department of Computer ScienceThe University of ManchesterManchesterUK
| | - Carole Goble
- Department of Computer ScienceThe University of ManchesterManchesterUK
| | - Xiaoming Hu
- Heidelberg Institute for Theoretical Studies (HITS)HeidelbergGermany
| | - Rupert W Overall
- German Center for Neurodegenerative Diseases (DZNE) DresdenDresdenGermany
- Center for Regenerative Therapies Dresden (CRTD)Technische Universität DresdenDresdenGermany
- Institute for BiologyHumboldt University of BerlinBerlinGermany
| | | | | | - Benjamin M Gyori
- Harvard Medical SchoolLaboratory of Systems PharmacologyBostonMAUSA
| | - John A Bachman
- Harvard Medical SchoolLaboratory of Systems PharmacologyBostonMAUSA
| | - Carlos Vega
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Valentin Grouès
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | | | - Pablo Porras
- European Bioinformatics Institute (EMBL‐EBI)European Molecular Biology LaboratoryHinxton, CambridgeshireUK
| | - Luana Licata
- Department of BiologyUniversity of Rome Tor VergataRomeItaly
| | | | - Francesca Sacco
- Department of BiologyUniversity of Rome Tor VergataRomeItaly
| | | | | | | | - Denes Turei
- Institute for Computational BiomedicineHeidelberg UniversityHeidelbergGermany
| | - Augustin Luna
- cBio Center, Divisions of Biostatistics and Computational BiologyDepartment of Data SciencesDana‐Farber Cancer InstituteBostonMAUSA
- Department of Cell BiologyHarvard Medical SchoolBostonMAUSA
| | - Ozgun Babur
- Computer Science DepartmentUniversity of Massachusetts BostonBostonMAUSA
| | | | - Alberto Valdeolivas
- Institute for Computational BiomedicineHeidelberg UniversityHeidelbergGermany
| | - Marina Esteban‐Medina
- Clinical Bioinformatics AreaFundación Progreso y Salud (FPS)Hospital Virgen del RocioSevillaSpain
- Computational Systems Medicine GroupInstitute of Biomedicine of Seville (IBIS)Hospital Virgen del RocioSevillaSpain
| | - Maria Peña‐Chilet
- Clinical Bioinformatics AreaFundación Progreso y Salud (FPS)Hospital Virgen del RocioSevillaSpain
- Computational Systems Medicine GroupInstitute of Biomedicine of Seville (IBIS)Hospital Virgen del RocioSevillaSpain
- Bioinformatics in Rare Diseases (BiER)Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)FPS, Hospital Virgen del RocíoSevillaSpain
| | - Kinza Rian
- Clinical Bioinformatics AreaFundación Progreso y Salud (FPS)Hospital Virgen del RocioSevillaSpain
- Computational Systems Medicine GroupInstitute of Biomedicine of Seville (IBIS)Hospital Virgen del RocioSevillaSpain
| | - Tomáš Helikar
- Department of BiochemistryUniversity of Nebraska‐LincolnLincolnNEUSA
| | | | - Dezso Modos
- Quadram Institute BioscienceNorwichUK
- Earlham InstituteNorwichUK
| | - Agatha Treveil
- Quadram Institute BioscienceNorwichUK
- Earlham InstituteNorwichUK
| | - Marton Olbei
- Quadram Institute BioscienceNorwichUK
- Earlham InstituteNorwichUK
| | | | - Stephane Ballereau
- Cancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
| | - Aurélien Dugourd
- Institute for Computational BiomedicineHeidelberg UniversityHeidelbergGermany
- Institute of Experimental Medicine and Systems BiologyFaculty of Medicine, RWTHAachen UniversityAachenGermany
| | | | - Vincent Noël
- Institut CuriePSL Research UniversityParisFrance
- INSERMParisFrance
- MINES ParisTechPSL Research UniversityParisFrance
| | - Laurence Calzone
- Institut CuriePSL Research UniversityParisFrance
- INSERMParisFrance
- MINES ParisTechPSL Research UniversityParisFrance
| | - Chris Sander
- cBio Center, Divisions of Biostatistics and Computational BiologyDepartment of Data SciencesDana‐Farber Cancer InstituteBostonMAUSA
- Department of Cell BiologyHarvard Medical SchoolBostonMAUSA
| | - Emek Demir
- Department of Molecular and Medical GeneticsOregon Health & Sciences UniversityPortlandORUSA
| | | | - Tom C Freeman
- The Roslin InstituteUniversity of EdinburghEdinburghUK
| | - Franck Augé
- Sanofi R&DTranslational SciencesChilly‐MazarinFrance
| | | | - Jan Hasenauer
- Helmholtz Zentrum München – German Research Center for Environmental HealthInstitute of Computational BiologyNeuherbergGermany
- Interdisciplinary Research Unit Mathematics and Life SciencesUniversity of BonnBonnGermany
| | - Olaf Wolkenhauer
- Department of Systems Biology and BioinformaticsUniversity of RostockRostockGermany
| | - Egon L Wilighagen
- Department of Bioinformatics ‐ BiGCaTNUTRIMMaastricht UniversityMaastrichtThe Netherlands
| | - Alexander R Pico
- Institute of Data Science and BiotechnologyGladstone InstitutesSan FranciscoCAUSA
| | - Chris T Evelo
- Department of Bioinformatics ‐ BiGCaTNUTRIMMaastricht UniversityMaastrichtThe Netherlands
- Maastricht Centre for Systems Biology (MaCSBio)Maastricht UniversityMaastrichtThe Netherlands
| | - Marc E Gillespie
- MaRS CentreOntario Institute for Cancer ResearchTorontoONCanada
- St. John’s University College of Pharmacy and Health SciencesQueensNYUSA
| | - Lincoln D Stein
- MaRS CentreOntario Institute for Cancer ResearchTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
| | - Henning Hermjakob
- European Bioinformatics Institute (EMBL‐EBI)European Molecular Biology LaboratoryHinxton, CambridgeshireUK
| | | | | | - Joaquin Dopazo
- Clinical Bioinformatics AreaFundación Progreso y Salud (FPS)Hospital Virgen del RocioSevillaSpain
- Computational Systems Medicine GroupInstitute of Biomedicine of Seville (IBIS)Hospital Virgen del RocioSevillaSpain
- Bioinformatics in Rare Diseases (BiER)Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)FPS, Hospital Virgen del RocíoSevillaSpain
- FPS/ELIXIR‐esHospital Virgen del RocíoSevillaSpain
| | - Alfonso Valencia
- Barcelona Supercomputing Center (BSC)BarcelonaSpain
- Institució Catalana de Recerca i Estudis Avançats (ICREA)BarcelonaSpain
| | - Hiroaki Kitano
- Systems Biology InstituteTokyoJapan
- Okinawa Institute of Science and Technology Graduate SchoolOkinawaJapan
| | - Emmanuel Barillot
- Institut CuriePSL Research UniversityParisFrance
- INSERMParisFrance
- MINES ParisTechPSL Research UniversityParisFrance
| | - Charles Auffray
- Cancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
| | - Rudi Balling
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Reinhard Schneider
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | | |
Collapse
|
57
|
Zhang Y, Lim CU, Sikirzhytski V, Naderi A, Chatzistamou I, Kiaris H. Propensity to endoplasmic reticulum stress in deer mouse fibroblasts predicts skin inflammation and body weight gain. Dis Model Mech 2021; 14:272498. [PMID: 34661243 PMCID: PMC8543066 DOI: 10.1242/dmm.049113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/26/2021] [Indexed: 01/04/2023] Open
Abstract
The unfolded protein response (UPR) is involved in the pathogenesis of metabolic disorders, yet whether variations in the UPR among individuals influence the propensity for metabolic disease remains unexplored. Using outbred deer mice as a model, we show that the intensity of UPR in fibroblasts isolated early in life predicts the extent of body weight gain after high-fat diet (HFD) administration. Contrary to those with intense UPR, animals with moderate UPR in fibroblasts and therefore displaying compromised stress resolution did not gain body weight but developed inflammation, especially in the skin, after HFD administration. Fibroblasts emerged as potent modifiers of this differential responsiveness to HFD, as indicated by the comparison of the UPR profiles of fibroblasts responding to fatty acids in vitro, by correlation analyses between UPR and proinflammatory cytokine-associated transcriptomes, and by BiP (also known as HSPA5) immunolocalization in skin lesions from animals receiving HFD. These results suggest that the UPR operates as a modifier of an individual's propensity for body weight gain in a manner that, at least in part, involves the regulation of an inflammatory response by skin fibroblasts. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Youwen Zhang
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Chang-Uk Lim
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Vitali Sikirzhytski
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Asieh Naderi
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Ioulia Chatzistamou
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| | - Hippokratis Kiaris
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA.,Peromyscus Genetic Stock Center, University of South Carolina, Columbia, SC 29208, USA
| |
Collapse
|
58
|
Claudiani S, Mason CC, Milojkovic D, Bianchi A, Pellegrini C, Di Marco A, Fiol CR, Robinson M, Ponnusamy K, Mokretar K, Chowdhury A, Albert M, Reid AG, Deininger MW, Naresh K, Apperley JF, Khorashad JS. Carfilzomib Enhances the Suppressive Effect of Ruxolitinib in Myelofibrosis. Cancers (Basel) 2021; 13:cancers13194863. [PMID: 34638347 PMCID: PMC8507927 DOI: 10.3390/cancers13194863] [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: 08/30/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 11/16/2022] Open
Abstract
As the first FDA-approved tyrosine kinase inhibitor for treatment of patients with myelofibrosis (MF), ruxolitinib improves clinical symptoms but does not lead to eradication of the disease or significant reduction of the mutated allele burden. The resistance of MF clones against the suppressive action of ruxolitinib may be due to intrinsic or extrinsic mechanisms leading to activity of additional pro-survival genes or signalling pathways that function independently of JAK2/STAT5. To identify alternative therapeutic targets, we applied a pooled-shRNA library targeting ~5000 genes to a JAK2V617F-positive cell line under a variety of conditions, including absence or presence of ruxolitinib and in the presence of a bone marrow microenvironment-like culture medium. We identified several proteasomal gene family members as essential to HEL cell survival. The importance of these genes was validated in MF cells using the proteasomal inhibitor carfilzomib, which also enhanced lethality in combination with ruxolitinib. We also showed that proteasome gene expression is reduced by ruxolitinib in MF CD34+ cells and that additional targeting of proteasomal activity by carfilzomib enhances the inhibitory action of ruxolitinib in vitro. Hence, this study suggests a potential role for proteasome inhibitors in combination with ruxolitinib for management of MF patients.
Collapse
Affiliation(s)
- Simone Claudiani
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
| | - Clinton C. Mason
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, University of Utah, Salt Lake City, UT 84108, USA;
| | - Dragana Milojkovic
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
| | - Andrea Bianchi
- Department of Information Engineering, University of L’Aquila, 67100 L’Aquila, Italy; (A.B.); (A.D.M.)
| | - Cristina Pellegrini
- Department of Biotechnological and Applied Clinical Science, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Antinisca Di Marco
- Department of Information Engineering, University of L’Aquila, 67100 L’Aquila, Italy; (A.B.); (A.D.M.)
| | - Carme R. Fiol
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
| | - Mark Robinson
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
| | - Kanagaraju Ponnusamy
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
| | - Katya Mokretar
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
| | - Avirup Chowdhury
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
| | - Michael Albert
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
| | - Alistair G. Reid
- Molecular Pathology Unit, Liverpool University, Liverpool L7 8XP, UK;
| | - Michael W. Deininger
- Versiti Blood Research Institute, Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| | - Kikkeri Naresh
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
| | - Jane F. Apperley
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
| | - Jamshid S. Khorashad
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College, London W12 0NN, UK; (S.C.); (D.M.); (C.R.F.); (M.R.); (K.P.); (K.M.); (A.C.); (M.A.); (K.N.); (J.F.A.)
- Correspondence:
| |
Collapse
|
59
|
Benedetti R, Gilardini Montani MS, Romeo MA, Arena A, Santarelli R, D’Orazi G, Cirone M. Role of UPR Sensor Activation in Cell Death-Survival Decision of Colon Cancer Cells Stressed by DPE Treatment. Biomedicines 2021; 9:1262. [PMID: 34572447 PMCID: PMC8466673 DOI: 10.3390/biomedicines9091262] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/08/2021] [Accepted: 09/16/2021] [Indexed: 12/25/2022] Open
Abstract
Polyphenols have been shown to possess several beneficial properties, including properties involved in the prevention or treatment of cancer. Among these polyphenols, a leading role is played by dihydroxyphenylethanol (DPE), the most powerful antioxidant compound contained in the olive oil. DPE has been previously reported to induce endoplasmic reticulum (ER) stress and to reduce cell survival in colon cancer, one of the most common and aggressive cancers in developed countries. In this study, we further investigated the activation of UPR by DPE and explored the roles of the three UPR sensors, inositol-requiring enzyme (IRE) 1 alpha, protein kinase RNA-like endoplasmic reticulum kinase (PERK), and activating transcription factor (ATF6), in the cell death-survival decision of wt and mutp53 colon cancer cells and the underlying mechanisms involved. We also unveiled a new interplay between ATF6 and wt, as well as mutp53, which may have important implications in cancer therapy.
Collapse
Affiliation(s)
- Rossella Benedetti
- Department of Experimental Medicine, La Sapienza University of Rome, Viale Regina Elena 324, 00185 Rome, Italy; (R.B.); (M.S.G.M.); (M.A.R.); (A.A.); (R.S.)
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00185 Rome, Italy
| | - Maria Saveria Gilardini Montani
- Department of Experimental Medicine, La Sapienza University of Rome, Viale Regina Elena 324, 00185 Rome, Italy; (R.B.); (M.S.G.M.); (M.A.R.); (A.A.); (R.S.)
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00185 Rome, Italy
| | - Maria Anele Romeo
- Department of Experimental Medicine, La Sapienza University of Rome, Viale Regina Elena 324, 00185 Rome, Italy; (R.B.); (M.S.G.M.); (M.A.R.); (A.A.); (R.S.)
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00185 Rome, Italy
| | - Andrea Arena
- Department of Experimental Medicine, La Sapienza University of Rome, Viale Regina Elena 324, 00185 Rome, Italy; (R.B.); (M.S.G.M.); (M.A.R.); (A.A.); (R.S.)
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00185 Rome, Italy
| | - Roberta Santarelli
- Department of Experimental Medicine, La Sapienza University of Rome, Viale Regina Elena 324, 00185 Rome, Italy; (R.B.); (M.S.G.M.); (M.A.R.); (A.A.); (R.S.)
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00185 Rome, Italy
| | - Gabriella D’Orazi
- Department of Neurosciences, Imaging and Clinical Sciences, University “G. D’Annunzio”, 66013 Chieti, Italy;
- Unit of Cellular Networks, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Mara Cirone
- Department of Experimental Medicine, La Sapienza University of Rome, Viale Regina Elena 324, 00185 Rome, Italy; (R.B.); (M.S.G.M.); (M.A.R.); (A.A.); (R.S.)
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00185 Rome, Italy
| |
Collapse
|
60
|
Lau E, Lee C, Li B, Pierro A. Endoplasmic reticulum stress in the acute intestinal epithelial injury of necrotizing enterocolitis. Pediatr Surg Int 2021; 37:1151-1160. [PMID: 34117913 DOI: 10.1007/s00383-021-04929-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/10/2021] [Indexed: 03/07/2023]
Abstract
Endoplasmic reticulum (ER) is a dynamic organelle that has many functions including protein synthesis, lipid synthesis, and calcium metabolism. Any perturbation in the ER such as accumulation of unfolded or misfolded proteins in the ER lumen causes ER stress. ER stress has been implicated in many intestinal inflammatory diseases. However, the role of ER stress in acute intestinal epithelial injuries such as necrotizing enterocolitis in preterm neonates, remains incompletely understood. In this review, we introduce ER structure, functions and summarize the intracellular signaling pathways involved in unfolded protein response (UPR), a survival mechanism in which cells exert an adaptive function to restore homeostasis in the ER. However, intense and prolonged ER stress induces apoptotic response which results in apoptotic cell death. We also discuss and highlight recent advances that have improved our understanding of the molecular mechanisms that regulate the ER stress in acute intestinal epithelial injuries such as necrotizing enterocolitis (NEC). We focus on the role of ER stress in influencing gut homeostasis in the neonatal period and on the potential therapeutic interventions to alleviate ER stress-induced cell death in NEC.
Collapse
Affiliation(s)
- Ethan Lau
- Division of General and Thoracic Surgery, The Hospital for Sick Children, University of Toronto, 1526-555 University Ave, Toronto, ON, M5G 1X8, Canada
| | - Carol Lee
- Division of General and Thoracic Surgery, The Hospital for Sick Children, University of Toronto, 1526-555 University Ave, Toronto, ON, M5G 1X8, Canada
| | - Bo Li
- Division of General and Thoracic Surgery, The Hospital for Sick Children, University of Toronto, 1526-555 University Ave, Toronto, ON, M5G 1X8, Canada
| | - Agostino Pierro
- Division of General and Thoracic Surgery, The Hospital for Sick Children, University of Toronto, 1526-555 University Ave, Toronto, ON, M5G 1X8, Canada.
| |
Collapse
|
61
|
Cherqaoui B, Araujo LM, Glatigny S, Breban M. Axial spondyloarthritis: emerging drug targets. Expert Opin Ther Targets 2021; 25:633-644. [PMID: 34431431 DOI: 10.1080/14728222.2021.1973429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Axial spondyloarthritis (AxSpA) is an inflammatory disorder that affects the joints, entheses, and bone tissues and is sometimes associated with psoriasis, anterior uveitis, and gut inflammation. Its pathogenesis is not wholly understood and treatment strategies require optimization. Data concerning AxSpA pathogenesis support a critical role of abnormal CD4+ T cell differentiation and exacerbated type 3 immune response. This knowledge boosted the development of interleukin (IL)-17 and Janus kinase inhibitors for AxSpA treatment beyond tumor necrosis factor-α inhibition. AREAS COVERED Emerging drug targets in animal and cellular models and with phase-II clinical trials have been evaluated. We also reflect on key issues for preclinical and clinical research going forward. EXPERT OPINION Some of the most promising approaches include: (i) modulation of transforming growth factor-β family that could exert a specific role on bone formation; (ii) blockade of granulocyte-macrophage colony-stimulating factor that could reduce type 3 immune responses, and (iii) rebalancing of biased immune response by cytokines such as IL-2 or IL-27 that could favor anti-inflammatory response and sustained drug-free remission. Multiomics tools and artificial intelligence could contribute to identification of optimal targets and help stratify patients for the most appropriate treatment options.
Collapse
Affiliation(s)
- Bilade Cherqaoui
- Infection & Inflammation, Umr 1173, Inserm, UVSQ/Université Paris Saclay - 2, Avenue De La Source De La Bièvre, Montigny-le-Bretonneux, France.,Inflamex - Laboratory of Excellence, University of Paris, France
| | - Luiza M Araujo
- Infection & Inflammation, Umr 1173, Inserm, UVSQ/Université Paris Saclay - 2, Avenue De La Source De La Bièvre, Montigny-le-Bretonneux, France.,Inflamex - Laboratory of Excellence, University of Paris, France
| | - Simon Glatigny
- Infection & Inflammation, Umr 1173, Inserm, UVSQ/Université Paris Saclay - 2, Avenue De La Source De La Bièvre, Montigny-le-Bretonneux, France.,Inflamex - Laboratory of Excellence, University of Paris, France
| | - Maxime Breban
- Infection & Inflammation, Umr 1173, Inserm, UVSQ/Université Paris Saclay - 2, Avenue De La Source De La Bièvre, Montigny-le-Bretonneux, France.,Inflamex - Laboratory of Excellence, University of Paris, France.,Department of Rheumatology, Ambroise Paré Hospital, Ap-hp - 9, Avenue Charles De Gaulle, Boulogne, France
| |
Collapse
|
62
|
Sun B, Tan D, Pan D, Baker MR, Liang Z, Wang Z, Lei J, Liu S, Hu CY, Li QX. Dihydromyricetin Imbues Antiadipogenic Effects on 3T3-L1 Cells via Direct Interactions with 78-kDa Glucose-Regulated Protein. J Nutr 2021; 151:1717-1725. [PMID: 33830233 DOI: 10.1093/jn/nxab057] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/15/2021] [Accepted: 02/11/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Obesity is among the most serious public health problems worldwide, with few safe pharmaceutical interventions. Natural products have become an important source of potential anti-obesity therapeutics. Dihydromyricetin (DHM) exerts antidiabetic effects. The biochemical target of DHM, however, has been unknown. It is crucial to identify the biochemical target of DHM for elucidating its physiological function and therapeutic value. OBJECTIVES The objective of this study was to identify the biochemical target of DHM. METHODS An abundant antiadipogenic flavanonol was extracted from the herbal plant Ampelopsis grossedentata through bioassay-guided fractionation and characterized with high-resolution LC-MS and 1H and 13C nuclear magnetic resonance. Antiadipogenic experiments were done with mouse 3T3-L1 preadipocytes. A biochemical target of the chemical of interest was identified with drug affinity responsive target stability assay. Direct interactions between the chemical of interest and the protein target in vitro were predicted with molecular docking and subsequently confirmed with surface plasmon resonance. Expression levels of peroxisome proliferator-activated receptor γ (PPARγ), which is associated with 78-kDa glucose-regulated protein (GRP78), were measured with real-time qPCR. RESULTS DHM was isolated, purified, and structurally characterized. Cellular studies showed that DHM notably reduced intracellular oil droplet formation in 3T3-L1 cells with a median effective concentration of 294 μM (i.e., 94 μg/mL). DHM targeted the ATP binding site of GRP78, which is associated with adipogenesis. An equilibrium dissociation constant between DHM and GRP78 was 21.8 μM. In 3T3-L1 cells upon treatment with DHM at 50 μM (i.e., 16 μg/mL), the expression level of PPARγ was downregulated to 53.9% of the solvent vehicle control's level. CONCLUSIONS DHM targets GRP78 in vitro. DHM is able to reduce lipid droplet formation in 3T3-L1 cells through a mode of action that is plausibly associated with direct interactions between GRP78 and DHM, which is a step forward in determining potential applications of DHM as an anti-obesity agent.
Collapse
Affiliation(s)
- Binmei Sun
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, HI, USA.,College of Horticulture, South China Agricultural University, Guangzhou, China
| | - Deguan Tan
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, HI, USA.,Institute of Tropical Bioscience and Biotechnology, Ministry of Agriculture Key Laboratory of Tropical Crops Biology and Genetic Resources and Hainan Academy of Tropical Agricultural Resource, Chinese Academy of Tropical Agriculural Sciences, Haikou, China
| | - Dongjin Pan
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, HI, USA.,Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Margaret R Baker
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Zhibin Liang
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Zhizheng Wang
- Key Laboratory of Pesticide & Chemical Biology, Ministry of Education, College of Chemistry, Central China Normal University, Wuhan, China
| | - Jianjun Lei
- College of Horticulture, South China Agricultural University, Guangzhou, China
| | - Shaoqun Liu
- College of Horticulture, South China Agricultural University, Guangzhou, China
| | - Ching Yuan Hu
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Qing X Li
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, HI, USA
| |
Collapse
|
63
|
Condé R, Hernandez-Torres E, Claudio-Piedras F, Recio-Tótoro B, Maya-Maldonado K, Cardoso-Jaime V, Lanz-Mendoza H. Heat Shock Causes Lower Plasmodium Infection Rates in Anopheles albimanus. Front Immunol 2021; 12:584660. [PMID: 34248924 PMCID: PMC8264367 DOI: 10.3389/fimmu.2021.584660] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 06/08/2021] [Indexed: 11/14/2022] Open
Abstract
The immune response of Anopheles mosquitoes to Plasmodium invasion has been extensively studied and shown to be mediated mainly by the nitric oxide synthase (NOS), dual oxidase (DUOX), phenoloxidase (PO), and antimicrobial peptides activity. Here, we studied the correlation between a heat shock insult, transcription of immune response genes, and subsequent susceptibility to Plasmodium berghei infection in Anopheles albimanus. We found that transcript levels of many immune genes were drastically affected by the thermal stress, either positively or negatively. Furthermore, the transcription of genes associated with modifications of nucleic acid methylation was affected, suggesting an increment in both DNA and RNA methylation. The heat shock increased PO and NOS activity in the hemolymph, as well as the transcription of several immune genes. As consequence, we observed that heat shock increased the resistance of mosquitoes to Plasmodium invasion. The data provided here could help the understanding of infection transmission under the ever more common heat waves.
Collapse
Affiliation(s)
- Renaud Condé
- Centro de Investigaciones Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Mexico
| | - Erika Hernandez-Torres
- Centro de Investigaciones Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Mexico
| | - Fabiola Claudio-Piedras
- Centro de Investigaciones Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Mexico
| | - Benito Recio-Tótoro
- Centro de Investigaciones Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Mexico.,Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Krystal Maya-Maldonado
- Centro de Investigaciones Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Mexico
| | - Victor Cardoso-Jaime
- Centro de Investigaciones Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Mexico
| | - Humberto Lanz-Mendoza
- Centro de Investigaciones Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Mexico
| |
Collapse
|
64
|
Xu B, Zhou L, Chen Q, Zhang J, Huang L, Wang S, Ye Z, Ren X, Cai Y, Jensen LD, Chen W, Li X, Ju R. Role of VEGFR2 in Mediating Endoplasmic Reticulum Stress Under Glucose Deprivation and Determining Cell Death, Oxidative Stress, and Inflammatory Factor Expression. Front Cell Dev Biol 2021; 9:631413. [PMID: 34222224 PMCID: PMC8249873 DOI: 10.3389/fcell.2021.631413] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 03/24/2021] [Indexed: 11/13/2022] Open
Abstract
Retinal pigment epithelium (RPE), a postmitotic monolayer located between the neuroretina and choroid, supports the retina and is closely associated with vision loss diseases such as age-related macular degeneration (AMD) upon dysfunction. Although environmental stresses are known to play critical roles in AMD pathogenesis and the roles of other stresses have been well investigated, glucose deprivation, which can arise from choriocapillary flow voids, has yet to be fully explored. In this study, we examined the involvement of VEGFR2 in glucose deprivation-mediated cell death and the underlying mechanisms. We found that VEGFR2 levels are a determinant for RPE cell death, a critical factor for dry AMD, under glucose deprivation. RNA sequencing analysis showed that upon VEGFR2 knockdown under glucose starvation, endoplasmic reticulum (ER) stress and unfolded protein response (UPR) are reduced. Consistently, VEGFR2 overexpression increased ER stress under the same condition. Although VEGFR2 was less expressed compared to EGFR1 and c-Met in RPE cells, it could elicit a higher level of ER stress induced by glucose starvation. Finally, downregulated VEGFR2 attenuated the oxidative stress and inflammatory factor expression, two downstream targets of ER stress. Our study, for the first time, has demonstrated a novel role of VEGFR2 in RPE cells under glucose deprivation, thus providing valuable insights into the mechanisms of AMD pathogenesis and suggesting that VEGFR2 might be a potential therapeutic target for AMD prevention, which may impede its progression.
Collapse
Affiliation(s)
- Bohan Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Linbin Zhou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Qishan Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jianing Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Lijuan Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Shasha Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Zhimin Ye
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xiangrong Ren
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yu Cai
- Chengdu Aier Eye Hospital, Chengdu, China
| | - Lasse Dahl Jensen
- Division of Cardiovascular Medicine, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Weirong Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xuri Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Rong Ju
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
65
|
ER Stress, UPR Activation and the Inflammatory Response to Viral Infection. Viruses 2021; 13:v13050798. [PMID: 33946891 PMCID: PMC8146799 DOI: 10.3390/v13050798] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/22/2021] [Accepted: 04/28/2021] [Indexed: 12/19/2022] Open
Abstract
The response to invading pathogens such as viruses is orchestrated by pattern recognition receptor (PRR) and unfolded protein response (UPR) signaling, which intersects and converges in the activation of proinflammatory pathways and the release of cytokines and chemokines that harness the immune system in the attempt to clear microbial infection. Despite this protective intent, the inflammatory response, particularly during viral infection, may be too intense or last for too long, whereby it becomes the cause of organ or systemic diseases itself. This suggests that a better understanding of the mechanisms that regulate this complex process is needed in order to achieve better control of the side effects that inflammation may cause while potentiating its protective role. The use of specific inhibitors of the UPR sensors or PRRs or the downstream pathways activated by their signaling could offer the opportunity to reach this goal and improve the outcome of inflammation-based diseases associated with viral infections.
Collapse
|
66
|
Kurt-Jones EA, Dudek TE, Watanabe D, Mandell L, Che J, Zhou S, Cao L, Greenough T, Babcock GJ, Diaz F, Oh HS, Zhou C, Finberg RW, Knipe DM. Expression of SARS coronavirus 1 spike protein from a herpesviral vector induces innate immune signaling and neutralizing antibody responses. Virology 2021; 559:165-172. [PMID: 33930819 PMCID: PMC8058630 DOI: 10.1016/j.virol.2021.04.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/16/2021] [Accepted: 04/16/2021] [Indexed: 01/05/2023]
Abstract
SARS coronavirus 1 (SARS-CoV-1) causes a respiratory infection that can lead to acute respiratory distress characterized by inflammation and high levels of cytokines in the lung tissue. In this study we constructed a herpes simplex virus 1 replication-defective mutant vector expressing SARS-CoV-1 spike protein as a potential vaccine vector and to probe the effects of spike protein on host cells. The spike protein expressed from this vector is functional in that it localizes to the surface of infected cells and induces fusion of ACE2-expressing cells. In immunized mice, the recombinant vector induced antibodies that bind to spike protein in an ELISA assay and that show neutralizing activity. The spike protein expressed from this vector can induce the expression of cytokines in an ACE2-independent, MyD88-dependent process. These results argue that the SARS-CoV-1 spike protein intrinsically activates signaling pathways that induce cytokines and contribute directly to the inflammatory process of SARS.
Collapse
Affiliation(s)
- Evelyn A Kurt-Jones
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Timothy E Dudek
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Daisuke Watanabe
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Leisa Mandell
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Jenny Che
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Shenghua Zhou
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - LuCheng Cao
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Thomas Greenough
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Gregory J Babcock
- MassBiologics, University of Massachusetts Medical School, Boston, MA, USA
| | - Fernando Diaz
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Hyung Suk Oh
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Changhong Zhou
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Robert W Finberg
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - David M Knipe
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
67
|
JAK/STAT inhibitor therapy partially rescues the lipodystrophic autoimmune phenotype in Clec16a KO mice. Sci Rep 2021; 11:7372. [PMID: 33795715 PMCID: PMC8016875 DOI: 10.1038/s41598-021-86493-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 03/08/2021] [Indexed: 02/06/2023] Open
Abstract
CLEC16A is implicated in multiple autoimmune diseases. We generated an inducible whole-body knockout (KO), Clec16aΔUBC mice to address the role of CLEC16A loss of function. KO mice exhibited loss of adipose tissue and severe weight loss in response to defective autophagic flux and exaggerated endoplasmic reticulum (ER) stress and robust cytokine storm. KO mice were glucose tolerant and displayed a state of systemic inflammation with elevated antibody levels, including IgM, IgA, Ig2b and IgG3, significantly reduced circulating insulin levels in the presence of normal food consumption. Metabolic analysis revealed disturbances in the lipid profile, white adipose decreasing concomitantly with enhanced inflammatory response, and energy wasting. Mechanistically, endoplasmic reticulum (ER) stress triggers excessive hormone sensitive lipases (HSL) mediated lipolysis which contributes to adipose inflammation via activation of JAK-STAT, stress kinases (ERK1/2, P38, JNK), and release of multiple proinflammatory mediators. Treatment with a JAK-STAT inhibitor (tofacitinib) partially rescued the inflammatory lipodystrophic phenotype and improved survival of Clec16aΔUBC mice by silencing cytokine release and modulating ER stress, lipolysis, mitophagy and autophagy. These results establish a mechanistic link between CLEC16A, lipid metabolism and the immune system perturbations. In summary, our Clec16aΔUBC mouse model highlights multifaceted roles of Clec16a in normal physiology, including a novel target for weight regulation and mutation-induced pathophysiology.
Collapse
|
68
|
Poncet AF, Bosteels V, Hoffmann E, Chehade S, Rennen S, Huot L, Peucelle V, Maréchal S, Khalife J, Blanchard N, Janssens S, Marion S. The UPR sensor IRE1α promotes dendritic cell responses to control Toxoplasma gondii infection. EMBO Rep 2021; 22:e49617. [PMID: 33586853 DOI: 10.15252/embr.201949617] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 01/23/2023] Open
Abstract
The unfolded protein response (UPR) has emerged as a central regulator of immune cell responses in several pathologic contexts including infections. However, how intracellular residing pathogens modulate the UPR in dendritic cells (DCs) and thereby affect T cell-mediated immunity remains uncharacterized. Here, we demonstrate that infection of DCs with Toxoplasma gondii (T. gondii) triggers a unique UPR signature hallmarked by the MyD88-dependent activation of the IRE1α pathway and the inhibition of the ATF6 pathway. Induction of XBP1s controls pro-inflammatory cytokine secretion in infected DCs, while IRE1α promotes MHCI antigen presentation of secreted parasite antigens. In mice, infection leads to a specific activation of the IRE1α pathway, which is restricted to the cDC1 subset. Mice deficient for IRE1α and XBP1 in DCs display a severe susceptibility to T. gondii and succumb during the acute phase of the infection. This early mortality is correlated with increased parasite burden and a defect in splenic T-cell responses. Thus, we identify the IRE1α/XBP1s branch of the UPR as a key regulator of host defense upon T. gondii infection.
Collapse
Affiliation(s)
- Anaïs F Poncet
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Victor Bosteels
- Laboratory for ER stress and Inflammation, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Eik Hoffmann
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Sylia Chehade
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Sofie Rennen
- Laboratory for ER stress and Inflammation, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Ludovic Huot
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Véronique Peucelle
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Sandra Maréchal
- Laboratory for ER stress and Inflammation, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Jamal Khalife
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Nicolas Blanchard
- Centre de Physiopathologie Toulouse Purpan (CPTP), INSERM, CNRS, UPS, Université de Toulouse, Toulouse, France
| | - Sophie Janssens
- Laboratory for ER stress and Inflammation, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Sabrina Marion
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| |
Collapse
|
69
|
Smith JA. STING, the Endoplasmic Reticulum, and Mitochondria: Is Three a Crowd or a Conversation? Front Immunol 2021; 11:611347. [PMID: 33552072 PMCID: PMC7858662 DOI: 10.3389/fimmu.2020.611347] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/04/2020] [Indexed: 12/20/2022] Open
Abstract
The anti-viral pattern recognition receptor STING and its partnering cytosolic DNA sensor cGAS have been increasingly recognized to respond to self DNA in multiple pathologic settings including cancer and autoimmune disease. Endogenous DNA sources that trigger STING include damaged nuclear DNA in micronuclei and mitochondrial DNA (mtDNA). STING resides in the endoplasmic reticulum (ER), and particularly in the ER-mitochondria associated membranes. This unique location renders STING well poised to respond to intracellular organelle stress. Whereas the pathways linking mtDNA and STING have been addressed recently, the mechanisms governing ER stress and STING interaction remain more opaque. The ER and mitochondria share a close anatomic and functional relationship, with mutual production of, and inter-organelle communication via calcium and reactive oxygen species (ROS). This interdependent relationship has potential to both generate the essential ligands for STING activation and to regulate its activity. Herein, we review the interactions between STING and mitochondria, STING and ER, ER and mitochondria (vis-à-vis calcium and ROS), and the evidence for 3-way communication.
Collapse
Affiliation(s)
- Judith A Smith
- Department of Pediatrics and Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
70
|
Robinson CM, Talty A, Logue SE, Mnich K, Gorman AM, Samali A. An Emerging Role for the Unfolded Protein Response in Pancreatic Cancer. Cancers (Basel) 2021; 13:cancers13020261. [PMID: 33445669 PMCID: PMC7828145 DOI: 10.3390/cancers13020261] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 12/17/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common form of pancreatic cancer and one of the leading causes of cancer-associated deaths in the world. It is characterised by dismal response rates to conventional therapies. A major challenge in treatment strategies for PDAC is the presence of a dense stroma that surrounds the tumour cells, shielding them from treatment. This unique tumour microenvironment is fuelled by paracrine signalling between pancreatic cancer cells and supporting stromal cell types including the pancreatic stellate cells (PSC). While our molecular understanding of PDAC is improving, there remains a vital need to develop effective, targeted treatments. The unfolded protein response (UPR) is an elaborate signalling network that governs the cellular response to perturbed protein homeostasis in the endoplasmic reticulum (ER) lumen. There is growing evidence that the UPR is constitutively active in PDAC and may contribute to the disease progression and the acquisition of resistance to therapy. Given the importance of the tumour microenvironment and cytokine signalling in PDAC, and an emerging role for the UPR in shaping the tumour microenvironment and in the regulation of cytokines in other cancer types, this review explores the importance of the UPR in PDAC biology and its potential as a therapeutic target in this disease.
Collapse
Affiliation(s)
- Claire M. Robinson
- Apoptosis Research Centre, School of Natural Sciences, National University of Ireland, H91 W2TY Galway, Ireland; (C.M.R.); (A.T.); (K.M.); (A.M.G.)
| | - Aaron Talty
- Apoptosis Research Centre, School of Natural Sciences, National University of Ireland, H91 W2TY Galway, Ireland; (C.M.R.); (A.T.); (K.M.); (A.M.G.)
| | - Susan E. Logue
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada;
- Research Institute in Oncology and Hematology, Cancer Care Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Katarzyna Mnich
- Apoptosis Research Centre, School of Natural Sciences, National University of Ireland, H91 W2TY Galway, Ireland; (C.M.R.); (A.T.); (K.M.); (A.M.G.)
| | - Adrienne M. Gorman
- Apoptosis Research Centre, School of Natural Sciences, National University of Ireland, H91 W2TY Galway, Ireland; (C.M.R.); (A.T.); (K.M.); (A.M.G.)
| | - Afshin Samali
- Apoptosis Research Centre, School of Natural Sciences, National University of Ireland, H91 W2TY Galway, Ireland; (C.M.R.); (A.T.); (K.M.); (A.M.G.)
- Correspondence:
| |
Collapse
|
71
|
Dymkowska D. The involvement of autophagy in the maintenance of endothelial homeostasis: The role of mitochondria. Mitochondrion 2021; 57:131-147. [PMID: 33412335 DOI: 10.1016/j.mito.2020.12.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/22/2020] [Accepted: 12/30/2020] [Indexed: 02/06/2023]
Abstract
Endothelial mitochondria play important signaling roles critical for the regulation of various cellular processes, including calcium signaling, ROS generation, NO synthesis or inflammatory response. Mitochondrial stress or disturbances in mitochondrial function may participate in the development and/or progression of endothelial dysfunction and could precede vascular diseases. Vascular functions are also strictly regulated by properly functioning degradation machinery, including autophagy and mitophagy, and tightly coordinated by mitochondrial and endoplasmic reticulum responses to stress. Within this review, current knowledge related to the development of cardiovascular disorders and the importance of mitochondria, endoplasmic reticulum and degradation mechanisms in vascular endothelial functions are summarized.
Collapse
Affiliation(s)
- Dorota Dymkowska
- The Laboratory of Cellular Metabolism, Nencki Institute of Experimental Biology PAS, 3 Pasteur str. 02-093 Warsaw, Poland.
| |
Collapse
|
72
|
Nash P, Kerschbaumer A, Dörner T, Dougados M, Fleischmann RM, Geissler K, McInnes I, Pope JE, van der Heijde D, Stoffer-Marx M, Takeuchi T, Trauner M, Winthrop KL, de Wit M, Aletaha D, Baraliakos X, Boehncke WH, Emery P, Isaacs JD, Kremer J, Lee EB, Maksymowych WP, Voshaar M, Tam LS, Tanaka Y, van den Bosch F, Westhovens R, Xavier R, Smolen JS. Points to consider for the treatment of immune-mediated inflammatory diseases with Janus kinase inhibitors: a consensus statement. Ann Rheum Dis 2021; 80:71-87. [PMID: 33158881 PMCID: PMC7788060 DOI: 10.1136/annrheumdis-2020-218398] [Citation(s) in RCA: 158] [Impact Index Per Article: 52.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Janus kinase inhibitors (JAKi) have been approved for use in various immune-mediated inflammatory diseases. With five agents licensed, it was timely to summarise the current understanding of JAKi use based on a systematic literature review (SLR) on efficacy and safety. METHODS Existing data were evaluated by a steering committee and subsequently reviewed by a 29 person expert committee leading to the formulation of a consensus statement that may assist the clinicians, patients and other stakeholders once the decision is made to commence a JAKi. The committee included patients, rheumatologists, a gastroenterologist, a haematologist, a dermatologist, an infectious disease specialist and a health professional. The SLR informed the Task Force on controlled and open clinical trials, registry data, phase 4 trials and meta-analyses. In addition, approval of new compounds by, and warnings from regulators that were issued after the end of the SLR search date were taken into consideration. RESULTS The Task Force agreed on and developed four general principles and a total of 26 points for consideration which were grouped into six areas addressing indications, treatment dose and comedication, contraindications, pretreatment screening and risks, laboratory and clinical follow-up examinations, and adverse events. Levels of evidence and strengths of recommendations were determined based on the SLR and levels of agreement were voted on for every point, reaching a range between 8.8 and 9.9 on a 10-point scale. CONCLUSION The consensus provides an assessment of evidence for efficacy and safety of an important therapeutic class with guidance on issues of practical management.
Collapse
Affiliation(s)
- Peter Nash
- School of Medicine, Griffith University, Brisbane, Queensland, Australia
| | - Andreas Kerschbaumer
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, 1090 Vienna, Austria
| | - Thomas Dörner
- Dept. Med./Rheumatology and Clinical Immunology, Charite Univ. Hospital, Berlin, Germany
| | - Maxime Dougados
- Hopital Cochin, Rheumatology, Université Paris Descartes, Paris, France
| | - Roy M Fleischmann
- Department of Medicine, Southwestern University of Texas, Dallas, Texas, USA
| | | | - Iain McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Janet E Pope
- Medicine, Division of Rheumatology, The University of Western Ontario, London, Ontario, Canada
| | | | - Michaela Stoffer-Marx
- Section for Outcomes Research, Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | | | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| | | | - Maarten de Wit
- Medical Humanities, Amsterdam University Medical Centre, Amsterdam, Netherlands
| | - Daniel Aletaha
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, 1090 Vienna, Austria
| | | | | | - Paul Emery
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Leeds, UK
| | - John D Isaacs
- Musculoskeletal Research Group, Newcastle University, Newcastle upon Tyne, UK
| | - Joel Kremer
- Rheumatology, Albany Medical College, Albany, New York, USA
| | - Eun Bong Lee
- Internal Medicine, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
| | - Walter P Maksymowych
- Medicine, University of Alberta Faculty of Medicine and Dentistry, Edmonton, Alberta, Canada
| | - Marieke Voshaar
- Medical Humanities, Amsterdam University Medical Centre, Amsterdam, Netherlands
| | - Lai-Shan Tam
- Department of Medicine & Therapeutics, Chinese University of Hong Kong Shaw College, New Territories, Hong Kong
| | - Yoshiya Tanaka
- First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | | | | | - Ricardo Xavier
- Division of Rheumatology, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Josef S Smolen
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
73
|
Nash P, Kerschbaumer A, Dörner T, Dougados M, Fleischmann RM, Geissler K, McInnes I, Pope JE, van der Heijde D, Stoffer-Marx M, Takeuchi T, Trauner M, Winthrop KL, de Wit M, Aletaha D, Baraliakos X, Boehncke WH, Emery P, Isaacs JD, Kremer J, Lee EB, Maksymowych WP, Voshaar M, Tam LS, Tanaka Y, van den Bosch F, Westhovens R, Xavier R, Smolen JS. Points to consider for the treatment of immune-mediated inflammatory diseases with Janus kinase inhibitors: a consensus statement. Ann Rheum Dis 2021. [PMID: 33158881 DOI: 10.1136/annrheumdis2020-218580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
OBJECTIVES Janus kinase inhibitors (JAKi) have been approved for use in various immune-mediated inflammatory diseases. With five agents licensed, it was timely to summarise the current understanding of JAKi use based on a systematic literature review (SLR) on efficacy and safety. METHODS Existing data were evaluated by a steering committee and subsequently reviewed by a 29 person expert committee leading to the formulation of a consensus statement that may assist the clinicians, patients and other stakeholders once the decision is made to commence a JAKi. The committee included patients, rheumatologists, a gastroenterologist, a haematologist, a dermatologist, an infectious disease specialist and a health professional. The SLR informed the Task Force on controlled and open clinical trials, registry data, phase 4 trials and meta-analyses. In addition, approval of new compounds by, and warnings from regulators that were issued after the end of the SLR search date were taken into consideration. RESULTS The Task Force agreed on and developed four general principles and a total of 26 points for consideration which were grouped into six areas addressing indications, treatment dose and comedication, contraindications, pretreatment screening and risks, laboratory and clinical follow-up examinations, and adverse events. Levels of evidence and strengths of recommendations were determined based on the SLR and levels of agreement were voted on for every point, reaching a range between 8.8 and 9.9 on a 10-point scale. CONCLUSION The consensus provides an assessment of evidence for efficacy and safety of an important therapeutic class with guidance on issues of practical management.
Collapse
Affiliation(s)
- Peter Nash
- School of Medicine, Griffith University, Brisbane, Queensland, Australia
| | - Andreas Kerschbaumer
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, 1090 Vienna, Austria
| | - Thomas Dörner
- Dept. Med./Rheumatology and Clinical Immunology, Charite Univ. Hospital, Berlin, Germany
| | - Maxime Dougados
- Hopital Cochin, Rheumatology, Université Paris Descartes, Paris, France
| | - Roy M Fleischmann
- Department of Medicine, Southwestern University of Texas, Dallas, Texas, USA
| | | | - Iain McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Janet E Pope
- Medicine, Division of Rheumatology, The University of Western Ontario, London, Ontario, Canada
| | | | - Michaela Stoffer-Marx
- Section for Outcomes Research, Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | | | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| | | | - Maarten de Wit
- Medical Humanities, Amsterdam University Medical Centre, Amsterdam, Netherlands
| | - Daniel Aletaha
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, 1090 Vienna, Austria
| | | | | | - Paul Emery
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Leeds, UK
| | - John D Isaacs
- Musculoskeletal Research Group, Newcastle University, Newcastle upon Tyne, UK
| | - Joel Kremer
- Rheumatology, Albany Medical College, Albany, New York, USA
| | - Eun Bong Lee
- Internal Medicine, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
| | - Walter P Maksymowych
- Medicine, University of Alberta Faculty of Medicine and Dentistry, Edmonton, Alberta, Canada
| | - Marieke Voshaar
- Medical Humanities, Amsterdam University Medical Centre, Amsterdam, Netherlands
| | - Lai-Shan Tam
- Department of Medicine & Therapeutics, Chinese University of Hong Kong Shaw College, New Territories, Hong Kong
| | - Yoshiya Tanaka
- First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | | | | | - Ricardo Xavier
- Division of Rheumatology, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Josef S Smolen
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
74
|
Ramalingam V, Rajaram R. A paradoxical role of reactive oxygen species in cancer signaling pathway: Physiology and pathology. Process Biochem 2021. [DOI: 10.1016/j.procbio.2020.09.032] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
75
|
Li M, Xie Y, Zhao K, Chen K, Cao Y, Zhang J, Han M, Hu L, He R, Wang D, Li H. Endoplasmic reticulum stress exacerbates inflammation in chronic rhinosinusitis with nasal polyps via the transcription factor XBP1. Clin Immunol 2020; 223:108659. [PMID: 33352294 DOI: 10.1016/j.clim.2020.108659] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/17/2020] [Accepted: 12/17/2020] [Indexed: 12/13/2022]
Abstract
Endoplasmic reticulum (ER) stress results in the activation of the unfolded protein response (UPR), a process that is involved in the pathogenesis of many inflammatory diseases. However, the role of ER stress in chronic rhinosinusitis with nasal polyps (CRSwNP) has yet to be elucidated. In this study, we found that the protein expression levels of a range of ER stress regulators, including p-PERK, ATF4, ATF6 and XBP1s, were significantly increased in CRSwNP compared to controls. Importantly, the expression of ATF4 and XBP1s was positively correlated with heightened inflammation in CRSwNP. In human nasal epithelial cells, the ER stress inducer tunicamycin (TM) could potentiate Toll-like receptors (TLRs) induced proinflammatory cytokines production. Furthermore, we found that the silencing of XBP1, but not ATF4 or ATF6, abrogated the proinflammatory effect of TM. Mechanistically, ER stress did not affect the NF-κB, MAPK or IRF3 signaling pathways. However, the ER stress regulator XBP1s was able to bind directly to the promoter region of inflammatory genes to modulate gene transcription. Besides, the commensal bacteria Staphylococcus aureus and several inflammatory factors, such as IL4, IL13, IL17 and IFNγ, could induce ER stress in epithelial cells. Collectively, ER stress plays a crucial role in facilitating TLR-induced inflammation. Targeting XBP1 can inhibit the inflammatory response, thus offering a potential approach to treat CRSwNP.
Collapse
Affiliation(s)
- Min Li
- ENT institute and Department of otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Yadong Xie
- ENT institute and Department of otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Keqing Zhao
- ENT institute and Department of otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Kun Chen
- Department of otorhinolaryngology, Xinhua hospital of Shanghai Jiao Tong University, Shanghai, China
| | - Yujie Cao
- ENT institute and Department of otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Jia Zhang
- ENT institute and Department of otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Miaomiao Han
- ENT institute and Department of otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Li Hu
- ENT institute and Department of otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Rui He
- Department of Immunology, MOE & MOH Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Dehui Wang
- ENT institute and Department of otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China.
| | - Huabin Li
- ENT institute and Department of otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
76
|
Perrotta C, Cattaneo MG, Molteni R, De Palma C. Autophagy in the Regulation of Tissue Differentiation and Homeostasis. Front Cell Dev Biol 2020; 8:602901. [PMID: 33363161 PMCID: PMC7758408 DOI: 10.3389/fcell.2020.602901] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/20/2020] [Indexed: 12/14/2022] Open
Abstract
Autophagy is a constitutive pathway that allows the lysosomal degradation of damaged components. This conserved process is essential for metabolic plasticity and tissue homeostasis and is crucial for mammalian post-mitotic cells. Autophagy also controls stem cell fate and defective autophagy is involved in many pathophysiological processes. In this review, we focus on established and recent breakthroughs aimed at elucidating the impact of autophagy in differentiation and homeostasis maintenance of endothelium, muscle, immune system, and brain providing a suitable framework of the emerging results and highlighting the pivotal role of autophagic response in tissue functions, stem cell dynamics and differentiation rates.
Collapse
Affiliation(s)
- Cristiana Perrotta
- Department of Biomedical and Clinical Sciences "Luigi Sacco" (DIBIC), Università degli Studi di Milano, Milan, Italy
| | - Maria Grazia Cattaneo
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, Milan, Italy
| | - Raffaella Molteni
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, Milan, Italy
| | - Clara De Palma
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
77
|
Sharip A, Kunz J. Understanding the Pathogenesis of Spondyloarthritis. Biomolecules 2020; 10:biom10101461. [PMID: 33092023 PMCID: PMC7588965 DOI: 10.3390/biom10101461] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 02/07/2023] Open
Abstract
Spondyloarthritis comprises a group of inflammatory diseases of the joints and spine, with various clinical manifestations. The group includes ankylosing spondylitis, reactive arthritis, psoriatic arthritis, arthritis associated with inflammatory bowel disease, and undifferentiated spondyloarthritis. The exact etiology and pathogenesis of spondyloarthritis are still unknown, but five hypotheses explaining the pathogenesis exist. These hypotheses suggest that spondyloarthritis is caused by arthritogenic peptides, an unfolded protein response, HLA-B*27 homodimer formation, malfunctioning endoplasmic reticulum aminopeptidases, and, last but not least, gut inflammation and dysbiosis. Here we discuss the five hypotheses and the evidence supporting each. In all of these hypotheses, HLA-B*27 plays a central role. It is likely that a combination of these hypotheses, with HLA-B*27 taking center stage, will eventually explain the development of spondyloarthritis in predisposed individuals.
Collapse
MESH Headings
- Arthritis, Psoriatic/genetics
- Arthritis, Psoriatic/immunology
- Arthritis, Psoriatic/metabolism
- Arthritis, Psoriatic/pathology
- Arthritis, Reactive/genetics
- Arthritis, Reactive/immunology
- Arthritis, Reactive/metabolism
- Arthritis, Reactive/pathology
- HLA-B27 Antigen/genetics
- HLA-B27 Antigen/immunology
- Humans
- Inflammation/genetics
- Inflammation/immunology
- Inflammation/metabolism
- Inflammation/pathology
- Inflammatory Bowel Diseases/genetics
- Inflammatory Bowel Diseases/immunology
- Inflammatory Bowel Diseases/metabolism
- Inflammatory Bowel Diseases/pathology
- Joints/immunology
- Joints/pathology
- Spine/immunology
- Spine/pathology
- Spondylarthritis/genetics
- Spondylarthritis/immunology
- Spondylarthritis/metabolism
- Spondylarthritis/pathology
- Spondylitis, Ankylosing/genetics
- Spondylitis, Ankylosing/immunology
- Spondylitis, Ankylosing/metabolism
- Spondylitis, Ankylosing/pathology
- Unfolded Protein Response/genetics
- Unfolded Protein Response/immunology
Collapse
|
78
|
Proteostasis Disturbances and Inflammation in Neurodegenerative Diseases. Cells 2020; 9:cells9102183. [PMID: 32998318 PMCID: PMC7601929 DOI: 10.3390/cells9102183] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/21/2020] [Accepted: 09/24/2020] [Indexed: 12/11/2022] Open
Abstract
Protein homeostasis (proteostasis) disturbances and inflammation are evident in normal aging and some age-related neurodegenerative diseases. While the proteostasis network maintains the integrity of intracellular and extracellular functional proteins, inflammation is a biological response to harmful stimuli. Cellular stress conditions can cause protein damage, thus exacerbating protein misfolding and leading to an eventual overload of the degradation system. The regulation of proteostasis network is particularly important in postmitotic neurons due to their limited regenerative capacity. Therefore, maintaining balanced protein synthesis, handling unfolding, refolding, and degrading misfolded proteins are essential to preserve all cellular functions in the central nervous sysytem. Failing proteostasis may trigger inflammatory responses in glial cells, and the consequent release of inflammatory mediators may lead to disturbances in proteostasis. Here, we review the mechanisms of proteostasis and inflammatory response, emphasizing their role in the pathological hallmarks of neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. Furthermore, we discuss the interplay between proteostatic stress and excessive immune response that activates inflammation and leads to dysfunctional proteostasis.
Collapse
|
79
|
Tanaka T, Warner BM, Odani T, Ji Y, Mo YQ, Nakamura H, Jang SI, Yin H, Michael DG, Hirata N, Suizu F, Ishigaki S, Oliveira FR, Motta ACF, Ribeiro-Silva A, Rocha EM, Atsumi T, Noguchi M, Chiorini JA. LAMP3 induces apoptosis and autoantigen release in Sjögren's syndrome patients. Sci Rep 2020; 10:15169. [PMID: 32939030 PMCID: PMC7494869 DOI: 10.1038/s41598-020-71669-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 08/10/2020] [Indexed: 12/16/2022] Open
Abstract
Primary Sjögren's syndrome (pSS) is a complex autoimmune disease characterized by dysfunction of secretory epithelia with only palliative therapy. Patients present with a constellation of symptoms, and the diversity of symptomatic presentation has made it difficult to understand the underlying disease mechanisms. In this study, aggregation of unbiased transcriptome profiling data sets of minor salivary gland biopsies from controls and Sjögren's syndrome patients identified increased expression of lysosome-associated membrane protein 3 (LAMP3/CD208/DC-LAMP) in a subset of Sjögren's syndrome cases. Stratification of patients based on their clinical characteristics suggested an association between increased LAMP3 expression and the presence of serum autoantibodies including anti-Ro/SSA, anti-La/SSB, anti-nuclear antibodies. In vitro studies demonstrated that LAMP3 expression induces epithelial cell dysfunction leading to apoptosis. Interestingly, LAMP3 expression resulted in the accumulation and release of intracellular TRIM21 (one component of SSA), La (SSB), and α-fodrin protein, common autoantigens in Sjögren's syndrome, via extracellular vesicles in an apoptosis-independent mechanism. This study defines a clear role for LAMP3 in the initiation of apoptosis and an independent pathway for the extracellular release of known autoantigens leading to the formation of autoantibodies associated with this disease.ClinicalTrials.gov Identifier: NCT00001196, NCT00001390, NCT02327884.
Collapse
Affiliation(s)
- Tsutomu Tanaka
- National Institute of Dental and Craniofacial Research, National Institutes of Health, NIH 10 Center Dr., Bethesda, MD, 20892, USA
| | - Blake M Warner
- National Institute of Dental and Craniofacial Research, National Institutes of Health, NIH 10 Center Dr., Bethesda, MD, 20892, USA
| | - Toshio Odani
- National Institute of Dental and Craniofacial Research, National Institutes of Health, NIH 10 Center Dr., Bethesda, MD, 20892, USA
| | - Youngmi Ji
- National Institute of Dental and Craniofacial Research, National Institutes of Health, NIH 10 Center Dr., Bethesda, MD, 20892, USA
| | - Ying-Qian Mo
- National Institute of Dental and Craniofacial Research, National Institutes of Health, NIH 10 Center Dr., Bethesda, MD, 20892, USA
| | - Hiroyuki Nakamura
- National Institute of Dental and Craniofacial Research, National Institutes of Health, NIH 10 Center Dr., Bethesda, MD, 20892, USA
| | - Shyh-Ing Jang
- National Institute of Dental and Craniofacial Research, National Institutes of Health, NIH 10 Center Dr., Bethesda, MD, 20892, USA
| | - Hongen Yin
- National Institute of Dental and Craniofacial Research, National Institutes of Health, NIH 10 Center Dr., Bethesda, MD, 20892, USA
| | - Drew G Michael
- National Institute of Dental and Craniofacial Research, National Institutes of Health, NIH 10 Center Dr., Bethesda, MD, 20892, USA
| | - Noriyuki Hirata
- Division of Cancer Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Futoshi Suizu
- Division of Cancer Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Satoko Ishigaki
- Division of Cancer Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Fabiola Reis Oliveira
- Department of Clinical Medicine, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Ana Carolina F Motta
- Department of Stomatology, Public Health and Forensic Dentistry, School of Dentistry of Ribeirão Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Alfredo Ribeiro-Silva
- Department of Pathology and Legal Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Eduardo M Rocha
- Department of Ophthalmology, Otorhinolaryngology, Head and Neck Surgery, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Tatsuya Atsumi
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masayuki Noguchi
- Division of Cancer Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - John A Chiorini
- National Institute of Dental and Craniofacial Research, National Institutes of Health, NIH 10 Center Dr., Bethesda, MD, 20892, USA.
| |
Collapse
|
80
|
Clayton PT. Is susceptibility to severe COVID-19 disease an inborn error of metabolism? J Inherit Metab Dis 2020; 43:906-907. [PMID: 32608016 PMCID: PMC7361907 DOI: 10.1002/jimd.12280] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 06/26/2020] [Accepted: 06/27/2020] [Indexed: 01/01/2023]
Affiliation(s)
- Peter T. Clayton
- Genetics and Genomic MedicineUCL Great Ormond Street Institute of Child HealthLondonUK
| |
Collapse
|
81
|
COVID-19 and Inflammatory Bowel Diseases: Risk Assessment, Shared Molecular Pathways, and Therapeutic Challenges. Gastroenterol Res Pract 2020; 2020:1918035. [PMID: 32714386 PMCID: PMC7352130 DOI: 10.1155/2020/1918035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 06/29/2020] [Indexed: 12/20/2022] Open
Abstract
Background The novel coronavirus SARS-CoV-2 causing COVID-19 disease is yielding a global outbreak with severe threats to public health. In this paper, we aimed at reviewing the current knowledge about COVID-19 infectious risk status in inflammatory bowel disease (IBD) patients requiring immunosuppressive medication. We also focused on several molecular insights that could explain why IBD patients appear not to have higher risks of infection and worse outcomes in COVID-19 than the general population in an attempt to provide scientific support for safer decisions in IBD patient care. Methods PubMed electronic database was interrogated for relevant articles involving data about common molecular pathways and shared treatment strategies between SARS-CoV-2, SARS-CoV-1, MERS-CoV, and inflammatory bowel diseases. Besides, Neural Covidex, an artificial intelligence tool, was used to answer queries about pathogenic coronaviruses and possible IBD interactions using the COVID-19 Open Research Dataset (CORD-19). Discussions. Few molecular and therapeutic interactions between IBD and pathogenic coronaviruses were explored. First, we showed how the activity of soluble angiotensin-converting enzyme 2, CD209L other receptors, and phosphorylated α subunit of eukaryotic translation initiation factor 2 might exert protective impact in IBD in case of coronavirus infection. Second, IBD medication was discussed in the context of possible beneficial effects on COVID-19 pathogeny, including “cytokine storm” prevention and treatment, immunomodulation, interferon signaling blocking, and viral endocytosis inhibition. Conclusions Using the current understanding of SARS-CoV-2 as well as other pathogenic coronaviruses immunopathology, we showed why IBD patients should not be considered at an increased risk of infection or more severe outcomes. Whether our findings are entirely applicable to the pathogenesis, disease susceptibility, and treatment management of SARS-CoV-2 infection in IBD must be further explored.
Collapse
|
82
|
Kuss-Duerkop SK, Keestra-Gounder AM. NOD1 and NOD2 Activation by Diverse Stimuli: a Possible Role for Sensing Pathogen-Induced Endoplasmic Reticulum Stress. Infect Immun 2020; 88:e00898-19. [PMID: 32229616 PMCID: PMC7309630 DOI: 10.1128/iai.00898-19] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Prompt recognition of microbes by cells is critical to eliminate invading pathogens. Some cell-associated pattern recognition receptors (PRRs) recognize and respond to microbial ligands. However, others can respond to cellular perturbations, such as damage-associated molecular patterns (DAMPs). Nucleotide oligomerization domains 1 and 2 (NOD1/2) are PRRs that recognize and respond to multiple stimuli of microbial and cellular origin, such as bacterial peptidoglycan, viral infections, parasitic infections, activated Rho GTPases, and endoplasmic reticulum (ER) stress. How NOD1/2 are stimulated by such diverse stimuli is not fully understood but may partly rely on cellular changes during infection that result in ER stress. NOD1/2 are ER stress sensors that facilitate proinflammatory responses for pathogen clearance; thus, NOD1/2 may help mount broad antimicrobial responses through detection of ER stress, which is often induced during a variety of infections. Some pathogens may subvert this response to promote infection through manipulation of NOD1/2 responses to ER stress that lead to apoptosis. Here, we review NOD1/2 stimuli and cellular responses. Furthermore, we discuss pathogen-induced ER stress and how it might potentiate NOD1/2 signaling.
Collapse
Affiliation(s)
- Sharon K Kuss-Duerkop
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - A Marijke Keestra-Gounder
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
83
|
Impact of Endoplasmic Reticulum Stress in Otorhinolaryngologic Diseases. Int J Mol Sci 2020; 21:ijms21114121. [PMID: 32527008 PMCID: PMC7312870 DOI: 10.3390/ijms21114121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/08/2020] [Accepted: 06/08/2020] [Indexed: 12/24/2022] Open
Abstract
The endoplasmic reticulum (ER) is an important organelle for normal cellular function and homeostasis in most living things. ER stress, which impairs ER function, occurs when the ER is overwhelmed by newly introduced immature proteins or when calcium in the ER is depleted. A number of diseases are associated with ER stress, including otorhinolaryngological diseases. The relationship between ER stress and otorhinolaryngologic conditions has been the subject of investigation over the last decade. Among otologic diseases associated with ER stress are otitis media and hearing loss. In rhinologic diseases, chronic rhinosinusitis, allergic rhinitis, and obstructive sleep apnea are also significantly associated with ER stress. In this review, we provide a comprehensive overview of the relationship between ER stress and otorhinolaryngological diseases, focusing on the current state of knowledge and mechanisms that link ER stress and otorhinolaryngologic diseases.
Collapse
|
84
|
KSHV infection skews macrophage polarisation towards M2-like/TAM and activates Ire1 α-XBP1 axis up-regulating pro-tumorigenic cytokine release and PD-L1 expression. Br J Cancer 2020; 123:298-306. [PMID: 32418990 PMCID: PMC7374093 DOI: 10.1038/s41416-020-0872-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 02/27/2020] [Accepted: 04/15/2020] [Indexed: 12/18/2022] Open
Abstract
Background Kaposi’s Sarcoma Herpesvirus (KSHV) is a gammaherpesvirus strongly linked to human cancer. The virus is also able to induce immune suppression, effect that contributes to onset/progression of the viral-associated malignancies. As KSHV may infect macrophages and these cells abundantly infiltrate Kaposi’s sarcoma lesions, in this study we investigated whether KSHV-infection could affect macrophage polarisation to promote tumorigenesis. Methods FACS analysis was used to detect macrophage markers and PD-L1 expression. KSHV infection and the molecular pathways activated were investigated by western blot analysis and by qRT-PCR while cytokine release was assessed by Multi-analyte Kit. Results We found that KSHV infection reduced macrophage survival and skewed their polarisation towards M2 like/TAM cells, based on the expression of CD163, on the activation of STAT3 and STAT6 pathways and the release of pro-tumorigenic cytokines such as IL-10, VEGF, IL-6 and IL-8. We also found that KSHV triggered Ire1 α-XBP1 axis activation in infected macrophages to increase the release of pro-tumorigenic cytokines and to up-regulate PD-L1 surface expression. Conclusions The findings that KSHV infection of macrophages skews their polarisation towards M2/TAM and that activate Ire1 α-XBP1 to increase the release of pro-tumorigenic cytokines and the expression of PD-L1, suggest that manipulation of UPR could be exploited to prevent or improve the treatment of KSHV-associated malignancies.
Collapse
|
85
|
Aguilar-Rojas A, Castellanos-Castro S, Matondo M, Gianetto QG, Varet H, Sismeiro O, Legendre R, Fernandes J, Hardy D, Coppée JY, Olivo-Marin JC, Guillen N. Insights into amebiasis using a human 3D-intestinal model. Cell Microbiol 2020; 22:e13203. [PMID: 32175652 DOI: 10.1111/cmi.13203] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 01/27/2020] [Accepted: 03/04/2020] [Indexed: 12/15/2022]
Abstract
Entamoeba histolytica is the causative agent of amebiasis, an infectious disease targeting the intestine and the liver in humans. Two types of intestinal infection are caused by this parasite: silent infection, which occurs in the majority of cases, and invasive disease, which affects 10% of infected persons. To understand the intestinal pathogenic process, several in vitro models, such as cell cultures, human tissue explants or human intestine xenografts in mice, have been employed. Nevertheless, our knowledge on the early steps of amebic intestinal infection and the molecules involved during human-parasite interaction is scarce, in part due to limitations in the experimental settings. In the present work, we took advantage of tissue engineering approaches to build a three-dimensional (3D)-intestinal model that is able to replicate the general characteristics of the human colon. This system consists of an epithelial layer that develops tight and adherens junctions, a mucus layer and a lamina propria-like compartment made up of collagen containing macrophages and fibroblast. By means of microscopy imaging, omics assays and the evaluation of immune responses, we show a very dynamic interaction between E. histolytica and the 3D-intestinal model. Our data highlight the importance of several virulence markers occurring in patients or in experimental models, but they also demonstrate the involvement of under described molecules and regulatory factors in the amoebic invasive process.
Collapse
Affiliation(s)
- Arturo Aguilar-Rojas
- Institut Pasteur, Bioimage Analysis Unit, Paris, France.,Instituto Mexicano del Seguro Social, Unidad de Investigación Médica en Medicina Reproductiva, Ciudad de México, Mexico
| | - Silvia Castellanos-Castro
- Institut Pasteur, Bioimage Analysis Unit, Paris, France.,Universidad Autónoma de la Ciudad de México, Colegio de Ciencias y Humanidades, Ciudad de México, Mexico
| | - Mariette Matondo
- Institut Pasteur, Plateforme Protéomique, Unité de Spectrométrie de Masse pour la Biologie (MSBio), Centrede Ressources et Recherches Technologiques (C2RT), Paris, France
| | - Quentin Giai Gianetto
- Institut Pasteur, Plateforme Protéomique, Unité de Spectrométrie de Masse pour la Biologie (MSBio), Centrede Ressources et Recherches Technologiques (C2RT), Paris, France.,Institut Pasteur, Plate-forme Transcriptome et EpiGenome, Biomics, Centre de Ressources et Recherches Technologiques (C2RT), Paris, France
| | - Hugo Varet
- Institut Pasteur, Plate-forme Transcriptome et EpiGenome, Biomics, Centre de Ressources et Recherches Technologiques (C2RT), Paris, France.,Institut Pasteur, Hub Bioinformatique et Biostatistique, Département de Biologie Computationnelle (USR3756 IP CNRS), Paris, France
| | - Odile Sismeiro
- Institut Pasteur, Plate-forme Transcriptome et EpiGenome, Biomics, Centre de Ressources et Recherches Technologiques (C2RT), Paris, France
| | - Rachel Legendre
- Institut Pasteur, Plate-forme Transcriptome et EpiGenome, Biomics, Centre de Ressources et Recherches Technologiques (C2RT), Paris, France.,Institut Pasteur, Hub Bioinformatique et Biostatistique, Département de Biologie Computationnelle (USR3756 IP CNRS), Paris, France
| | - Julien Fernandes
- Institut Pasteur, UTechSPBI, Centre de Ressources et Recherches Technologiques (C2RT), Paris, France
| | - David Hardy
- Institut Pasteur, Experimental Neuropathology Unit, Paris, France
| | - Jean-Yves Coppée
- Institut Pasteur, Plate-forme Transcriptome et EpiGenome, Biomics, Centre de Ressources et Recherches Technologiques (C2RT), Paris, France
| | | | - Nancy Guillen
- Institut Pasteur, Paris, France.,Centre National de la Recherche Scientifique, Paris, France
| |
Collapse
|
86
|
Raiter A, Lipovetzki J, Lubin I, Yerushalmi R. GRP78 expression in peripheral blood mononuclear cells is a new predictive marker for the benefit of taxanes in breast cancer neoadjuvant treatment. BMC Cancer 2020; 20:333. [PMID: 32306920 PMCID: PMC7168854 DOI: 10.1186/s12885-020-06835-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 04/06/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Breast cancer treatment is tailored to the specific cancer subtype. Often, systemic treatment is given prior to surgery. Chemotherapy induces significant endoplasmic reticulum (ER) stress-mediated cell death and upregulation of 78-kDa glucose-regulated protein (GRP78). We hypothesized that chemotherapy induces ER stress not only in the tumor tissue but also in immune cells, which may affect the response to anti-cancer treatment. METHODS We determined the surface expression of GRP78 on 15 different peripheral blood mononuclear cell (PBMC) subpopulations in 20 breast cancer patients at three time points of the neoadjuvant treatment, i.e., at baseline, after anthracycline treatment, and after taxanes treatment. For this purpose, we performed flow cytometric analyses and analyzed the data using ANOVA and the Tukey test. Serum cytokine levels were also evaluated, and their levels were correlated with response to treatment using the t-test after log transformation and Mann-Whitney U Wilcoxon W test. RESULTS A significant increase in GRP78 expression in PBMCs was documented during the taxane phase, only in patients who achieved pathological complete response (pCR). GRP78-positive clones correlated with increased serum levels of interferon gamma (IFNγ). CONCLUSIONS The presence of GRP78-positive clones in certain PBMC subpopulations in pCR patients suggests a dynamic interaction between ER stress and immune responsiveness. The correlation of GRP78-positive clones with increased levels of IFNγ supports the idea that GRP78 expression in PBMCs might serve as a new predictive marker to identify the possible benefits of taxanes in the neoadjuvant setting.
Collapse
Affiliation(s)
- Annat Raiter
- Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Rabin Medical Center, Beilinson Campus, 49100, Petach Tikva, Israel.
| | - Julia Lipovetzki
- Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Rabin Medical Center, Beilinson Campus, 49100, Petach Tikva, Israel
| | - Ido Lubin
- Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Rabin Medical Center, Beilinson Campus, 49100, Petach Tikva, Israel
| | - Rinat Yerushalmi
- Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Rabin Medical Center, Beilinson Campus, 49100, Petach Tikva, Israel.
- Davidoff Cancer Center, Rabin Medical Center, Beilinson Campus, 49100, Petach Tikva, Israel.
| |
Collapse
|
87
|
Salminen A, Kaarniranta K, Kauppinen A. ER stress activates immunosuppressive network: implications for aging and Alzheimer's disease. J Mol Med (Berl) 2020; 98:633-650. [PMID: 32279085 PMCID: PMC7220864 DOI: 10.1007/s00109-020-01904-z] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/24/2020] [Accepted: 03/26/2020] [Indexed: 12/14/2022]
Abstract
The endoplasmic reticulum (ER) contains stress sensors which recognize the accumulation of unfolded proteins within the lumen of ER, and subsequently these transducers stimulate the unfolded protein response (UPR). The ER sensors include the IRE1, PERK, and ATF6 transducers which activate the UPR in an attempt to restore the quality of protein folding and thus maintain cellular homeostasis. If there is excessive stress, UPR signaling generates alarmins, e.g., chemokines and cytokines, which activate not only tissue-resident immune cells but also recruit myeloid and lymphoid cells into the affected tissues. ER stress is a crucial inducer of inflammation in many pathological conditions. A chronic low-grade inflammation and cellular senescence have been associated with the aging process and many age-related diseases, such as Alzheimer’s disease. Currently, it is known that immune cells can exhibit great plasticity, i.e., they are able to display both pro-inflammatory and anti-inflammatory phenotypes in a context-dependent manner. The microenvironment encountered in chronic inflammatory conditions triggers a compensatory immunosuppression which defends tissues from excessive inflammation. Recent studies have revealed that chronic ER stress augments the suppressive phenotypes of immune cells, e.g., in tumors and other inflammatory disorders. The activation of immunosuppressive network, including myeloid-derived suppressor cells (MDSC) and regulatory T cells (Treg), has been involved in the aging process and Alzheimer’s disease. We will examine in detail whether the ER stress-related changes found in aging tissues and Alzheimer’s disease are associated with the activation of immunosuppressive network, as has been observed in tumors and many chronic inflammatory diseases.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.,Department of Ophthalmology, Kuopio University Hospital, P.O. Box 100, FI-70029, Kuopio, Finland
| | - Anu Kauppinen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| |
Collapse
|
88
|
Tong Y, Yang L, Shao F, Yan X, Li X, Huang G, Xiao Y, Zhou Z. Distinct secretion pattern of serum proinsulin in different types of diabetes. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:452. [PMID: 32395496 PMCID: PMC7210169 DOI: 10.21037/atm.2020.03.189] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Background Latent autoimmune diabetes in adults (LADA) is characterized by autoimmunity, late-onset and intermediate beta-cell deprivation rate between type 2 diabetes mellitus (T2DM) and type 1 diabetes mellitus (T1DM). Herein, we investigated proinsulin (PI) secretion patterns and the endoplasmic reticulum (ER) dysfunction biomarker, PI-to-C-peptide (PI:CP) ratio, to elucidate beta-cell intrinsic pathogenesis mechanisms in different types of diabetes. Methods Total serum fasting PI (FPI) were measured in adult-onset and newly-diagnosed diabetes patients, including 60 T1DM, 60 LADA and 60 T2DM. Thirty of each type underwent mixed meal tolerance tests (MMTTs), and hence 120 min postprandial PI (PPI) were detected. PI:CP ratio = PI (pmol/L) ÷ CP (pmol/L) × 100%. PI-related measurements among types of diabetes were compared. Correlation between PI-related measurements and beta-cell autoimmunity were analyzed. The possibility of discriminating LADA from T1DM and T2DM with PI-related measurements were tested. Results FPI and PPI were significantly higher in LADA than T1DM (P<0.001 for both comparisons), but lower than those in T2DM (P<0.001 and P=0.026, respectively). Fasting PI:CP ratio was significantly higher in T1DM than both LADA and T2DM (median 3.25% vs. 2.13% and 2.32%, P=0.011 and P=0.017, respectively). In LADA, positive autoantibody numbers increased by both fasting and postprandial PI:CP ratio (P=0.007 and P=0.034, respectively). Areas under receiver operation characteristic curves (AUCROC) of FPI and PPI for discriminating LADA from adult-onset T1DM were 0.751 (P<0.001) and 0.838 (P<0.001), respectively. Between LADA and T2DM, AUCROC of FPI and PPI were 0.685 (P<0.001) and 0.741 (P=0.001), respectively. Conclusions In the development of autoimmune diabetes, interplays between ER stress and beta-cell autoimmunity are potentially responsible for severer beta-cell destruction. PI-related measurements could help in differentiating LADA from adult-onset T1DM and T2DM.
Collapse
Affiliation(s)
- Yue Tong
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha 410011, China.,National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.,Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, Changsha 410011, China
| | - Lin Yang
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha 410011, China.,National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.,Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, Changsha 410011, China
| | - Feng Shao
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha 410011, China.,National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.,Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, Changsha 410011, China
| | - Xiang Yan
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha 410011, China.,National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.,Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, Changsha 410011, China
| | - Xia Li
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha 410011, China.,National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.,Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, Changsha 410011, China
| | - Gan Huang
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha 410011, China.,National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.,Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, Changsha 410011, China
| | - Yang Xiao
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha 410011, China.,National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.,Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, Changsha 410011, China
| | - Zhiguang Zhou
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha 410011, China.,National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.,Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, Changsha 410011, China
| |
Collapse
|
89
|
Serrano-Del Valle A, Naval J, Anel A, Marzo I. Novel Forms of Immunomodulation for Cancer Therapy. Trends Cancer 2020; 6:518-532. [PMID: 32460005 DOI: 10.1016/j.trecan.2020.02.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/07/2020] [Accepted: 02/19/2020] [Indexed: 02/07/2023]
Abstract
In recent years immunotherapy has provided new hope for cancer patients. However, some patients eventually relapse. Immunological responses are thought to underlie the long-term effects of conventional or targeted therapies. Whether this influence emerges from direct effects on cancer cells through immunogenic cell death (ICD) or by modulating the immune environment requires further clarification. ICD-related molecular mechanisms are also shared by cell-intrinsic defense responses that combat foreign intrusions. Indeed, we could potentially mimic and harness these processes to improve cancer immunogenicity. In addition, the microbiome is materializing as a missing factor in the cancer-immune therapy axis. The emerging idea of manipulating the gut microbiota to improve responses to anticancer therapy is becoming increasingly popular, but further clinical authentication is needed.
Collapse
Affiliation(s)
- Alfonso Serrano-Del Valle
- Apoptosis, Immunity, and Cancer Group, Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, and Aragon Health Research Institute (IIS-Aragon), Zaragoza 50009, Spain.
| | - Javier Naval
- Apoptosis, Immunity, and Cancer Group, Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, and Aragon Health Research Institute (IIS-Aragon), Zaragoza 50009, Spain
| | - Alberto Anel
- Apoptosis, Immunity, and Cancer Group, Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, and Aragon Health Research Institute (IIS-Aragon), Zaragoza 50009, Spain
| | - Isabel Marzo
- Apoptosis, Immunity, and Cancer Group, Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, and Aragon Health Research Institute (IIS-Aragon), Zaragoza 50009, Spain
| |
Collapse
|
90
|
Chamberlain N, Anathy V. Pathological consequences of the unfolded protein response and downstream protein disulphide isomerases in pulmonary viral infection and disease. J Biochem 2020; 167:173-184. [PMID: 31790139 PMCID: PMC6988748 DOI: 10.1093/jb/mvz101] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 11/08/2019] [Indexed: 12/15/2022] Open
Abstract
Protein folding within the endoplasmic reticulum (ER) exists in a delicate balance; perturbations of this balance can overload the folding capacity of the ER and disruptions of ER homoeostasis is implicated in numerous diseases. The unfolded protein response (UPR), a complex adaptive stress response, attempts to restore normal proteostasis, in part, through the up-regulation of various foldases and chaperone proteins including redox-active protein disulphide isomerases (PDIs). There are currently over 20 members of the PDI family each consisting of varying numbers of thioredoxin-like domains which, generally, assist in oxidative folding and disulphide bond rearrangement of peptides. While there is a large amount of redundancy in client proteins of the various PDIs, the size of the family would indicate more nuanced roles for the individual PDIs. However, the role of individual PDIs in disease pathogenesis remains uncertain. The following review briefly discusses recent findings of ER stress, the UPR and the role of individual PDIs in various respiratory disease states.
Collapse
Affiliation(s)
- Nicolas Chamberlain
- Department of Pathology and Laboratory Medicine, University of Vermont Larner College of Medicine, 149 Beaumont Ave, Burlington, VT 05405, USA
| | - Vikas Anathy
- Department of Pathology and Laboratory Medicine, University of Vermont Larner College of Medicine, 149 Beaumont Ave, Burlington, VT 05405, USA
| |
Collapse
|
91
|
Fania L, Provini A, Salemme A, Sinagra JL, Guerra L, Mazzanti C, Didona B, Castiglia D, Di Zenzo G. Development of bullous pemphigoid in junctional epidermolysis bullosa. J Eur Acad Dermatol Venereol 2020; 34:e146-e148. [PMID: 31709656 DOI: 10.1111/jdv.16057] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- L Fania
- First Dermatologic Division, IDI-IRCCS, Rome, Italy
| | - A Provini
- First Dermatologic Division, IDI-IRCCS, Rome, Italy
| | - A Salemme
- Laboratory of Molecular and Cell Biology, IDI-IRCCS, Rome, Italy
| | - J L Sinagra
- Laboratory of Molecular and Cell Biology, IDI-IRCCS, Rome, Italy
| | - L Guerra
- Laboratory of Molecular and Cell Biology, IDI-IRCCS, Rome, Italy
| | - C Mazzanti
- First Dermatologic Division, IDI-IRCCS, Rome, Italy
| | - B Didona
- First Dermatologic Division, IDI-IRCCS, Rome, Italy
| | - D Castiglia
- Laboratory of Molecular and Cell Biology, IDI-IRCCS, Rome, Italy
| | - G Di Zenzo
- Laboratory of Molecular and Cell Biology, IDI-IRCCS, Rome, Italy
| |
Collapse
|
92
|
Murugina NE, Budikhina AS, Dagil YA, Maximchik PV, Balyasova LS, Murugin VV, Melnikov MV, Sharova VS, Nikolaeva AM, Chkadua GZ, Pinegin BV, Pashenkov MV. Glycolytic reprogramming of macrophages activated by NOD1 and TLR4 agonists: No association with proinflammatory cytokine production in normoxia. J Biol Chem 2020; 295:3099-3114. [PMID: 32005665 DOI: 10.1074/jbc.ra119.010589] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 01/20/2020] [Indexed: 12/13/2022] Open
Abstract
Upon activation with pathogen-associated molecular patterns, metabolism of macrophages and dendritic cells is shifted from oxidative phosphorylation to aerobic glycolysis, which is considered important for proinflammatory cytokine production. Fragments of bacterial peptidoglycan (muramyl peptides) activate innate immune cells through nucleotide-binding oligomerization domain (NOD) 1 and/or NOD2 receptors. Here, we show that NOD1 and NOD2 agonists induce early glycolytic reprogramming of human monocyte-derived macrophages (MDM), which is similar to that induced by the Toll-like receptor 4 (TLR4) agonist lipopolysaccharide. This glycolytic reprogramming depends on Akt kinases, independent of mTOR complex 1 and is efficiently inhibited by 2-deoxy-d-glucose (2-DG) or by glucose starvation. 2-DG inhibits proinflammatory cytokine production by MDM and monocyte-derived dendritic cells activated by NOD1 or TLR4 agonists, except for tumor necrosis factor production by MDM, which is inhibited initially, but augmented 4 h after addition of agonists and later. However, 2-DG exerts these effects by inducing unfolded protein response rather than by inhibiting glycolysis. By contrast, glucose starvation does not cause unfolded protein response and, in normoxic conditions, only marginally affects proinflammatory cytokine production triggered through NOD1 or TLR4. In hypoxia mimicked by treating MDM with oligomycin (a mitochondrial ATP synthase inhibitor), both 2-DG and glucose starvation strongly suppress tumor necrosis factor and interleukin-6 production and compromise cell viability. In summary, the requirement of glycolytic reprogramming for proinflammatory cytokine production in normoxia is not obvious, and effects of 2-DG on cytokine responses should be interpreted cautiously. In hypoxia, however, glycolysis becomes critical for cytokine production and cell survival.
Collapse
Affiliation(s)
- Nina E Murugina
- Laboratory of Clinical Immunology, National Research Center, Institute of Immunology, Federal Medical-Biological Agency of Russia, Kashirskoe shosse 24, 115522 Moscow, Russia
| | - Anna S Budikhina
- Laboratory of Clinical Immunology, National Research Center, Institute of Immunology, Federal Medical-Biological Agency of Russia, Kashirskoe shosse 24, 115522 Moscow, Russia
| | - Yulia A Dagil
- Laboratory of Clinical Immunology, National Research Center, Institute of Immunology, Federal Medical-Biological Agency of Russia, Kashirskoe shosse 24, 115522 Moscow, Russia
| | - Polina V Maximchik
- Faculty of Fundamental Medicine, Lomonosov Moscow State University, Leninskie Gory 1, 119991 Moscow, Russia
| | - Lyudmila S Balyasova
- Laboratory of Clinical Immunology, National Research Center, Institute of Immunology, Federal Medical-Biological Agency of Russia, Kashirskoe shosse 24, 115522 Moscow, Russia
| | - Vladimir V Murugin
- Laboratory of Clinical Immunology, National Research Center, Institute of Immunology, Federal Medical-Biological Agency of Russia, Kashirskoe shosse 24, 115522 Moscow, Russia
| | - Mikhail V Melnikov
- Laboratory of Clinical Immunology, National Research Center, Institute of Immunology, Federal Medical-Biological Agency of Russia, Kashirskoe shosse 24, 115522 Moscow, Russia; Department of Neurology, Neurosurgery and Medical Genetics, Pirogov Russian National Research Medical University, Ostrovityanova street 1, 117997 Moscow, Russia
| | - Viktoriya S Sharova
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Vavilova street 26, 119334 Moscow, Russia
| | - Anna M Nikolaeva
- Laboratory of Clinical Immunology, National Research Center, Institute of Immunology, Federal Medical-Biological Agency of Russia, Kashirskoe shosse 24, 115522 Moscow, Russia; Biological Faculty, Lomonosov Moscow State University, Leninskie Gory 1, 119991 Moscow, Russia
| | - Georgy Z Chkadua
- Laboratory of Experimental Diagnostics and Biotherapy of Tumors, N. N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of the Russian Federation, Kashirskoe shosse 24 Building 2, 115522 Moscow, Russia
| | - Boris V Pinegin
- Laboratory of Clinical Immunology, National Research Center, Institute of Immunology, Federal Medical-Biological Agency of Russia, Kashirskoe shosse 24, 115522 Moscow, Russia
| | - Mikhail V Pashenkov
- Laboratory of Clinical Immunology, National Research Center, Institute of Immunology, Federal Medical-Biological Agency of Russia, Kashirskoe shosse 24, 115522 Moscow, Russia.
| |
Collapse
|
93
|
Hell L, Lurger K, Mauracher LM, Grilz E, Reumiller CM, Schmidt GJ, Ercan H, Koder S, Assinger A, Basilio J, Gebhart J, Ay C, Pabinger I, Zellner M. Altered platelet proteome in lupus anticoagulant (LA)-positive patients-protein disulfide isomerase and NETosis as new players in LA-related thrombosis. Exp Mol Med 2020; 52:66-78. [PMID: 31956273 PMCID: PMC7000701 DOI: 10.1038/s12276-019-0358-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 09/06/2019] [Accepted: 09/25/2019] [Indexed: 01/22/2023] Open
Abstract
Patients with antiphospholipid syndrome (APS) are at high risk of developing venous and arterial thromboembolism (TE). The role of platelets in the pathogenesis of these prothrombotic conditions is not yet fully understood. The aim of this study was to gain mechanistic insights into the role of platelets in APS by comparing the platelet proteome between lupus anticoagulant (LA)-positive patients with (LA+ TE+) and without a history of TE (LA+ TE-) and healthy controls. The platelet proteome of 47 patients with LA, 31 with a history of TE and 16 without thrombotic history, and 47 healthy controls was analyzed by two-dimensional differential in-gel electrophoresis and mass spectrometry to identify disease-related proteins. Afterward, selected LA-related platelet proteins were validated by western blot and ELISA. Alterations of 25 proteins were observed between the study groups. STRING pathway analysis showed that LA-related protein profiles were involved in platelet activation, aggregation, and degranulation. For example, protein disulfide isomerase family members, enzymes that promote thrombosis, were upregulated in platelets and plasma of LA+ TE+ patients. Leukocyte elastase inhibitor (SERPINB1), an antagonist of neutrophil extracellular trap (NET) formation, was decreased in platelets of LA+ TE+ patients compared to healthy controls. Additionally, citrullinated histone H3, a NET-specific marker, was increased in plasma of LA+ TE+ patients. These findings suggest that decreased platelet SERPINB1 levels favor prothrombotic NETosis, especially in LA+ TE+ patients. Our findings reveal protein abundance changes connected to altered platelet function in LA-positive patients, thus suggesting a pathogenic role of platelets in thrombotic complications in APS.
Collapse
Affiliation(s)
- Lena Hell
- 0000 0000 9259 8492grid.22937.3dClinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Kristina Lurger
- 0000 0000 9259 8492grid.22937.3dClinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Lisa-Marie Mauracher
- 0000 0000 9259 8492grid.22937.3dClinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Ella Grilz
- 0000 0000 9259 8492grid.22937.3dClinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Christina Maria Reumiller
- 0000 0000 9259 8492grid.22937.3dCenter for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Georg Johannes Schmidt
- 0000 0000 9259 8492grid.22937.3dDepartment of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Huriye Ercan
- 0000 0000 9259 8492grid.22937.3dClinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Silvia Koder
- 0000 0000 9259 8492grid.22937.3dClinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Alice Assinger
- 0000 0000 9259 8492grid.22937.3dCenter for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - José Basilio
- 0000 0000 9259 8492grid.22937.3dCenter for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Johanna Gebhart
- 0000 0000 9259 8492grid.22937.3dClinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Cihan Ay
- 0000 0000 9259 8492grid.22937.3dClinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Ingrid Pabinger
- 0000 0000 9259 8492grid.22937.3dClinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Maria Zellner
- 0000 0000 9259 8492grid.22937.3dCenter for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
94
|
Laviada-Molina HA, Leal-Berumen I, Rodriguez-Ayala E, Bastarrachea RA. Working Hypothesis for Glucose Metabolism and SARS-CoV-2 Replication: Interplay Between the Hexosamine Pathway and Interferon RF5 Triggering Hyperinflammation. Role of BCG Vaccine? Front Endocrinol (Lausanne) 2020; 11:514. [PMID: 32733388 PMCID: PMC7358362 DOI: 10.3389/fendo.2020.00514] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 06/26/2020] [Indexed: 12/20/2022] Open
Affiliation(s)
| | - Irene Leal-Berumen
- Facultad de Medicina y Ciencias Biomédicas, Universidad Autónoma de Chihuahua, Chihuahua, México
| | - Ernesto Rodriguez-Ayala
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Norte, Naucalpan de Juárez, México
| | - Raul A. Bastarrachea
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, United States
- *Correspondence: Raul A. Bastarrachea
| |
Collapse
|
95
|
Abstract
Etiologies of human pathophysiology have been associated with states of severe inflammation. The endothelium defender P53 supports cellular functions, by orchestrating anti-inflammatory responses. The purpose of the present article is to provide an update on the mechanisms enforcing the protective actions of P53 in human homeostasis, and to discuss current efforts on the development of new therapies against inflammatory abnormalities.
Collapse
Affiliation(s)
- Khadeja-Tul Kubra
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA
| | - Mohammad S Akhter
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA
| | - Mohammad A Uddin
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA
| | - Nektarios Barabutis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA
| |
Collapse
|
96
|
Nascimento Da Conceicao V, Sun Y, Zboril EK, De la Chapa JJ, Singh BB. Loss of Ca 2+ entry via Orai-TRPC1 induces ER stress, initiating immune activation in macrophages. J Cell Sci 2019; 133:jcs237610. [PMID: 31722977 PMCID: PMC10682644 DOI: 10.1242/jcs.237610] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 10/25/2019] [Indexed: 12/17/2022] Open
Abstract
Activation of cellular stresses is associated with inflammation; however, the mechanisms are not well identified. Here, we provide evidence that loss of Ca2+ influx induces endoplasmic reticulum (ER) stress in primary macrophages and in murine macrophage cell line Raw 264.7, in which the unfolded protein response is initiated to modulate cytokine production, thereby activating the immune response. Stressors that initiate the ER stress response block store-dependent Ca2+ entry in macrophages prior to the activation of the unfolded protein response. The endogenous Ca2+ entry channel is dependent on the Orai1-TRPC1-STIM1 complex, and the presence of ER stressors decreased expression of TRPC1, Orai1 and STIM1. Additionally, blocking Ca2+ entry with SKF96365 also induced ER stress, promoted cytokine production, activation of autophagy, increased caspase activation and induced apoptosis. Furthermore, ER stress inducers inhibited cell cycle progression, promoted the inflammatory M1 phenotype, and increased phagocytosis. Mechanistically, restoration of Orai1-STIM1 expression inhibited the ER stress-mediated loss of Ca2+ entry that prevents ER stress and inhibits cytokine production, and thus induced cell survival. These results suggest an unequivocal role of Ca2+ entry in modulating ER stress and in the induction of inflammation.
Collapse
Affiliation(s)
| | - Yuyang Sun
- Department of Periodontics, School of Dentistry, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Emily K Zboril
- Department of Periodontics, School of Dentistry, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Jorge J De la Chapa
- Department of Comprehensive Dentistry, School of Dentistry, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Brij B Singh
- Department of Periodontics, School of Dentistry, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
97
|
Studencka-Turski M, Çetin G, Junker H, Ebstein F, Krüger E. Molecular Insight Into the IRE1α-Mediated Type I Interferon Response Induced by Proteasome Impairment in Myeloid Cells of the Brain. Front Immunol 2019; 10:2900. [PMID: 31921161 PMCID: PMC6932173 DOI: 10.3389/fimmu.2019.02900] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 11/26/2019] [Indexed: 12/24/2022] Open
Abstract
Proteostasis is critical for cells to maintain the balance between protein synthesis, quality control, and degradation. This is particularly important for myeloid cells of the central nervous system as their immunological function relies on proper intracellular protein turnover by the ubiquitin-proteasome system. Accordingly, disruption of proteasome activity due to, e.g., loss-of-function mutations within genes encoding proteasome subunits, results in systemic autoinflammation. On the molecular level, pharmacological inhibition of proteasome results in endoplasmic reticulum (ER) stress-activated unfolded protein response (UPR) as well as an induction of type I interferons (IFN). Nevertheless, our understanding as to whether and to which extent UPR signaling regulates type I IFN response is limited. To address this issue, we have tested the effects of proteasome dysfunction upon treatment with proteasome inhibitors in primary murine microglia and microglia-like cell line BV-2. Our data show that proteasome impairment by bortezomib is a stimulus that activates all three intracellular ER-stress transducers activation transcription factor 6, protein kinase R-like endoplasmic reticulum kinase and inositol-requiring protein 1 alpha (IRE1α), causing a full activation of the UPR. We further demonstrate that impaired proteasome activity in microglia cells triggers an induction of IFNβ1 in an IRE1-dependent manner. An inhibition of the IRE1 endoribonuclease activity significantly attenuates TANK-binding kinase 1-mediated activation of type I IFN. Moreover, interfering with TANK-binding kinase 1 activity also compromised the expression of C/EBP homologous protein 10, thereby emphasizing a multilayered interplay between UPR and type IFN response pathway. Interestingly, the induced protein kinase R-like endoplasmic reticulum kinase-activation transcription factor 4-C/EBP homologous protein 10 and IRE1-X-box-binding protein 1 axes caused a significant upregulation of proinflammatory cytokine interleukin 6 expression that exacerbates STAT1/STAT3 signaling in cells with dysfunctional proteasomes. Altogether, these findings indicate that proteasome impairment disrupts ER homeostasis and triggers a complex interchange between ER-stress sensors and type I IFN signaling, thus inducing in myeloid cells a state of chronic inflammation.
Collapse
Affiliation(s)
- Maja Studencka-Turski
- Institute of Medical Biochemistry and Molecular Biology, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Gonca Çetin
- Institute of Medical Biochemistry and Molecular Biology, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Heike Junker
- Institute of Medical Biochemistry and Molecular Biology, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Frédéric Ebstein
- Institute of Medical Biochemistry and Molecular Biology, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Elke Krüger
- Institute of Medical Biochemistry and Molecular Biology, Universitätsmedizin Greifswald, Greifswald, Germany
| |
Collapse
|
98
|
Endoplasmic reticulum stress is involved in ventilator-induced lung injury in mice via the IRE1α-TRAF2-NF-κB pathway. Int Immunopharmacol 2019; 78:106069. [PMID: 31841755 DOI: 10.1016/j.intimp.2019.106069] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 12/12/2022]
Abstract
Inflammation plays a criticalrole in the development of ventilator-induced lung injury (VILI). Endoplasmic reticulum (ER) stress is associated with a variety of diseases through the modulation of inflammatory responses. However, little is known about how ER stress is implicated in VILI. In this study, murine mechanical ventilation models were constructed. Total protein and inflammatory cytokines were measured in bronchoalveolar lavage fluid (BALF),and lung tissue injurywasassessedby histology. Our data revealed that mice subjected to high tidal ventilation (TV) for 4 h showed more severe pulmonary edema and inflammation than those of mice with spontaneous breathing and low TV-treatment. In addition, the high TV-treated animals upregulated the ER stress markers GRP78, CHOP, p-IRE1α, TRAF2, and p-NF-κB expression at both the mRNA and protein levels in lung tissue. Administration of thapsigargin exacerbated the histological changes, inflammation and expression of GRP78 and CHOP after high TV, but treatment with ER stress and IRE1α kinase inhibitors attenuated the pathological damage and downregulated the high expression of GRP78, CHOP, p-IRE1α, TRAF2, and p-NF-κB, suggesting that ER stress is involved in VILI though the IRE1α/TRAF2/NF-κB signaling pathway in mice.
Collapse
|
99
|
Imai J, Otani M, Sakai T. Distinct Subcellular Compartments of Dendritic Cells Used for Cross-Presentation. Int J Mol Sci 2019; 20:ijms20225606. [PMID: 31717517 PMCID: PMC6888166 DOI: 10.3390/ijms20225606] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/29/2019] [Accepted: 11/06/2019] [Indexed: 02/06/2023] Open
Abstract
Dendritic cells (DCs) present exogenous protein-derived peptides on major histocompatibility complex class I molecules to prime naïve CD8+ T cells. This DC specific ability, called cross-presentation (CP), is important for the activation of cell-mediated immunity and the induction of self-tolerance. Recent research revealed that endoplasmic reticulum-associated degradation (ERAD), which was first identified as a part of the unfolded protein response—a quality control system in the ER—plays a pivotal role in the processing of exogenous proteins in CP. Moreover, DCs express a variety of immuno-modulatory molecules and cytokines to regulate T cell activation in response to the environment. Although both CP and immuno-modulation are indispensable, contrasting ER conditions are required for their correct activity. Since ERAD substrates are unfolded proteins, their accumulation may result in ER stress, impaired cell homeostasis, and eventually apoptosis. In contrast, activation of the unfolded protein response should be inhibited for DCs to express immuno-modulatory molecules and cytokines. Here, we review recent advances on antigen CP, focusing on intracellular transport routes for exogenous antigens and distinctive subcellular compartments involved in ERAD.
Collapse
Affiliation(s)
- Jun Imai
- Correspondence: ; Tel.: +81-27-352-1180
| | | | | |
Collapse
|
100
|
Sag D, Ayyildiz ZO, Gunalp S, Wingender G. The Role of TRAIL/DRs in the Modulation of Immune Cells and Responses. Cancers (Basel) 2019; 11:cancers11101469. [PMID: 31574961 PMCID: PMC6826877 DOI: 10.3390/cancers11101469] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/09/2019] [Accepted: 09/20/2019] [Indexed: 12/26/2022] Open
Abstract
Expression of TRAIL (tumor necrosis factor–related apoptosis–inducing ligand) by immune cells can lead to the induction of apoptosis in tumor cells. However, it becomes increasingly clear that the interaction of TRAIL and its death receptors (DRs) can also directly impact immune cells and influence immune responses. Here, we review what is known about the role of TRAIL/DRs in immune cells and immune responses in general and in the tumor microenvironment in particular.
Collapse
Affiliation(s)
- Duygu Sag
- Izmir Biomedicine and Genome Center (IBG), 35340 Balcova/Izmir, Turkey.
- Department of Medical Biology, Faculty of Medicine, Dokuz Eylul University, 35340 Balcova/Izmir, Turkey.
- Department of Genome Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Balcova/Izmir, Turkey.
| | - Zeynep Ozge Ayyildiz
- Department of Genome Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Balcova/Izmir, Turkey.
| | - Sinem Gunalp
- Department of Genome Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Balcova/Izmir, Turkey.
| | - Gerhard Wingender
- Izmir Biomedicine and Genome Center (IBG), 35340 Balcova/Izmir, Turkey.
- Department of Biomedicine and Health Technologies, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Balcova/Izmir, Turkey.
| |
Collapse
|