1
|
Ghosh S, Tuz AA, Stenzel M, Singh V, Richter M, Soehnlein O, Lange E, Heyer R, Cibir Z, Beer A, Jung M, Nagel D, Hermann DM, Hasenberg A, Grüneboom A, Sickmann A, Gunzer M. Proteomic characterization of 1,000 human and murine neutrophils freshly isolated from blood and sites of sterile inflammation. Mol Cell Proteomics 2024:100858. [PMID: 39395581 DOI: 10.1016/j.mcpro.2024.100858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/18/2024] [Accepted: 10/09/2024] [Indexed: 10/14/2024] Open
Abstract
Neutrophils are indispensable for defense against pathogens. Injured tissue-infiltrated neutrophils can establish a niche of chronic inflammation and promote degeneration. Studies investigated transcriptome of single-infiltrated neutrophils which could misinterpret molecular states of these post mitotic cells. However, neutrophil proteome characterization has been challenging due to low harvests from affected tissues. Here, we present a workflow to obtain proteome of 1,000 murine and human tissue-infiltrated neutrophils. We generated spectral libraries containing ∼6,200 mouse and ∼5,300 human proteins from circulating neutrophils. 4,800 mouse and 3,400 human proteins were recovered from 1,000 cells with 102-108 copies/cell. Neutrophils from stroke-affected mouse brains adapted to the glucose-deprived environment with increased mitochondrial activity and ROS-production while cells invading inflamed human oral cavities increased phagocytosis and granule release. We provide an extensive protein repository for resting human and mouse neutrophils, identify proteins lost in low input samples, thus enabling the proteomic characterization of limited tissue infiltrated neutrophils.
Collapse
Affiliation(s)
- Susmita Ghosh
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | - Ali Ata Tuz
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Martin Stenzel
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | - Vikramjeet Singh
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Mathis Richter
- Institute for Experimental Pathology, University of Münster, Germany
| | - Oliver Soehnlein
- Institute for Experimental Pathology, University of Münster, Germany
| | - Emanuel Lange
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | - Robert Heyer
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany; Multidimensional Omics Analyses Group, Faculty of Technology, Bielefeld University, Universitätsstraße 25, 33615 Bielefeld, Germany
| | - Zülal Cibir
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Alexander Beer
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Marcel Jung
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Dennis Nagel
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Dirk M Hermann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Anja Hasenberg
- Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Anika Grüneboom
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany; Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801, Bochum, Germany; Department of Chemistry, College of Physical Sciences, University of Aberdeen, AB24 3FX, Aberdeen, UK.
| | - Matthias Gunzer
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany; Institute for Experimental Immunology and Imaging, University Hospital, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
2
|
Sasvári P, Pettkó-Szandtner A, Wisniewski É, Csépányi-Kömi R. Neutrophil-specific interactome of ARHGAP25 reveals novel partners and regulatory insights. Sci Rep 2024; 14:20106. [PMID: 39210013 PMCID: PMC11362597 DOI: 10.1038/s41598-024-71002-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
ARHGAP25, a crucial molecule in immunological processes, serves as a Rac-specific GTPase-activating protein. Its role in cell migration and phagocyte functions, affecting the outcome of complex immunological diseases such as rheumatoid arthritis, renders it a promising target for drug research. Despite its importance, our knowledge of its intracellular interactions is still limited. This study employed proteomic analysis of glutathione S-transferase (GST)-tag pulldowns and co-immunoprecipitation from neutrophilic granulocyte cell lysate, revealing 76 candidates for potential physical interactions that complement ARHGAP25's known profile. Notably, four small GTPases (RAC2, RHOG, ARF4, and RAB27A) exhibited high affinity for ARHGAP25. The ARHGAP25-RAC2 and ARHGAP25-RHOG interactions appeared to be affected by the activation state of the small GTPases, suggesting a GTP-GDP cycle-dependent interaction. In silico dimer prediction pinpointed ARHGAP25's GAP domain as a credible binding interface, suggesting its suitability for GTP hydrolysis. Additionally, a list of Fc receptor-related kinases, phosphatases, and three of the 14-3-3 members were identified as potential partners, with in silico predictions highlighting eight binding sites, presenting novel insight on a potential regulatory mechanism for ARHGAP25.
Collapse
Affiliation(s)
- Péter Sasvári
- Department of Physiology, Semmelweis University, Tűzoltó u. 37-47., Budapest, 1085, Hungary
| | - Aladár Pettkó-Szandtner
- Proteomics Research Group, Core Facility, HUN-REN Biological Research Centre, Szeged, 6726, Hungary
| | - Éva Wisniewski
- Department of Physiology, Semmelweis University, Tűzoltó u. 37-47., Budapest, 1085, Hungary
| | - Roland Csépányi-Kömi
- Department of Physiology, Semmelweis University, Tűzoltó u. 37-47., Budapest, 1085, Hungary.
| |
Collapse
|
3
|
Naveh CA, Roberts K, Zakrzewski P, Rice CM, Ponce-Garcia FM, Fleming K, Thompson M, Panyapiean N, Jiang H, Diezmann S, Moura PL, Toye AM, Amulic B. Neutrophils cultured ex vivo from CD34 + stem cells are immature and genetically tractable. J Transl Med 2024; 22:526. [PMID: 38822352 PMCID: PMC11143668 DOI: 10.1186/s12967-024-05337-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 05/22/2024] [Indexed: 06/02/2024] Open
Abstract
BACKGROUND Neutrophils are granulocytes with essential antimicrobial effector functions and short lifespans. During infection or sterile inflammation, emergency granulopoiesis leads to release of immature neutrophils from the bone marrow, serving to boost circulating neutrophil counts. Steady state and emergency granulopoiesis are incompletely understood, partly due to a lack of genetically amenable models of neutrophil development. METHODS We optimised a method for ex vivo production of human neutrophils from CD34+ haematopoietic progenitors. Using flow cytometry, we phenotypically compared cultured neutrophils with native neutrophils from donors experiencing emergency granulopoiesis, and steady state neutrophils from non-challenged donors. We carry out functional and proteomic characterisation of cultured neutrophils and establish genome editing of progenitors. RESULTS We obtain high yields of ex vivo cultured neutrophils, which phenotypically resemble immature neutrophils released into the circulation during emergency granulopoiesis. Cultured neutrophils have similar rates of ROS production and bacterial killing but altered degranulation, cytokine release and antifungal activity compared to mature neutrophils isolated from peripheral blood. These differences are likely due to incomplete synthesis of granule proteins, as demonstrated by proteomic analysis. CONCLUSION Ex vivo cultured neutrophils are genetically tractable via genome editing of precursors and provide a powerful model system for investigating the properties and behaviour of immature neutrophils.
Collapse
Affiliation(s)
- Claire A Naveh
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Kiran Roberts
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Przemysław Zakrzewski
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Christopher M Rice
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Fernando M Ponce-Garcia
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Kathryn Fleming
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Megan Thompson
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Nawamin Panyapiean
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Huan Jiang
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Stephanie Diezmann
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Pedro L Moura
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge (MedH), Karolinska Institutet, Huddinge, Sweden
| | - Ashley M Toye
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK.
| | - Borko Amulic
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK.
| |
Collapse
|
4
|
Díaz-Varela M, Sanchez-Hidalgo A, Calderon-Copete S, Tacchini V, Shipley TR, Ramírez LG, Marquis J, Fernández OL, Saravia NG, Tacchini-Cottier F. The different impact of drug-resistant Leishmania on the transcription programs activated in neutrophils. iScience 2024; 27:109773. [PMID: 38711445 PMCID: PMC11070714 DOI: 10.1016/j.isci.2024.109773] [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: 01/09/2024] [Revised: 02/22/2024] [Accepted: 04/15/2024] [Indexed: 05/08/2024] Open
Abstract
Drug resistance threatens the effective control of infections, including parasitic diseases such as leishmaniases. Neutrophils are essential players in antimicrobial control, but their role in drug-resistant infections is poorly understood. Here, we evaluated human neutrophil response to clinical parasite strains having distinct natural drug susceptibility. We found that Leishmania antimony drug resistance significantly altered the expression of neutrophil genes, some of them transcribed by specific neutrophil subsets. Infection with drug-resistant parasites increased the expression of detoxification pathways and reduced the production of cytokines. Among these, the chemokine CCL3 was predominantly impacted, which resulted in an impaired ability of neutrophils to attract myeloid cells. Moreover, decreased myeloid recruitment when CCL3 levels are reduced was confirmed by blocking CCL3 in a mouse model. Collectively, these findings reveal that the interplay between naturally drug-resistant parasites and neutrophils modulates the infected skin immune microenvironment, revealing a key role of neutrophils in drug resistance.
Collapse
Affiliation(s)
- Míriam Díaz-Varela
- Department of Immunobiology, WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, 1066 Epalinges, Switzerland
| | - Andrea Sanchez-Hidalgo
- Centro Internacional de Entrenamiento e Investigaciones Médicas, CIDEIM, Cali 760031, Colombia
- Universidad Icesi, Cali 760031, Colombia
| | - Sandra Calderon-Copete
- Lausanne Genomic Technologies Facility, University of Lausanne, 1015 Lausanne, Switzerland
| | - Virginie Tacchini
- Department of Immunobiology, WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, 1066 Epalinges, Switzerland
| | - Tobias R. Shipley
- Department of Immunobiology, WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, 1066 Epalinges, Switzerland
| | - Lady Giovanna Ramírez
- Centro Internacional de Entrenamiento e Investigaciones Médicas, CIDEIM, Cali 760031, Colombia
- Universidad Icesi, Cali 760031, Colombia
| | - Julien Marquis
- Lausanne Genomic Technologies Facility, University of Lausanne, 1015 Lausanne, Switzerland
| | - Olga Lucía Fernández
- Centro Internacional de Entrenamiento e Investigaciones Médicas, CIDEIM, Cali 760031, Colombia
- Universidad Icesi, Cali 760031, Colombia
| | - Nancy Gore Saravia
- Centro Internacional de Entrenamiento e Investigaciones Médicas, CIDEIM, Cali 760031, Colombia
- Universidad Icesi, Cali 760031, Colombia
| | - Fabienne Tacchini-Cottier
- Department of Immunobiology, WHO Collaborative Center for Research and Training in Immunology, University of Lausanne, 1066 Epalinges, Switzerland
| |
Collapse
|
5
|
Li J, Ye LJ, Dai YW, Wang HW, Gao J, Shen YH, Wang F, Dai QG, Wu YQ. Single-cell analysis reveals a unique microenvironment in peri-implantitis. J Clin Periodontol 2024. [PMID: 38566468 DOI: 10.1111/jcpe.13982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 01/31/2024] [Accepted: 03/14/2024] [Indexed: 04/04/2024]
Abstract
AIM This study aimed to reveal the unique microenvironment of peri-implantitis through single-cell analysis. MATERIALS AND METHODS Herein, we performed single-cell RNA sequencing (scRNA-seq) of biopsies from patients with peri-implantitis (PI) and compared the results with healthy individuals (H) and patients with periodontitis (PD). RESULTS Decreased numbers of stromal cells and increased immune cells were found in the PI group, which implies a severe inflammatory infiltration. The fibroblasts were found to be heterogeneous and the specific pro-inflammatory CXCL13+ sub-cluster was more represented in the PI group, in contrast to the PD and H groups. Furthermore, more neutrophil infiltration was detected in the PI group than in the PD group, and cell-cell communication and ligand-receptor pairs revealed most neutrophils were recruited by CXCL13+ fibroblasts through CXCL8/CXCL6-CXCR2/CXCR1. Notably, our study demonstrated that the unique microenvironment of the PI group promoted the differentiation of monocyte/macrophage lineage cells into osteoclasts, which might explain the faster and more severe bone resorption in the progression of PI than PD. CONCLUSIONS Collectively, this study suggests a unique immune microenvironment of PI, which may explain the differences between PI and PD in the clinic. These outcomes will aid in finding new specific and effective treatments for PI.
Collapse
Affiliation(s)
- J Li
- Department of Second Dental Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - L J Ye
- Department of Second Dental Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Y W Dai
- Department of Second Dental Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - H W Wang
- Department of Second Dental Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - J Gao
- Department of Second Dental Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Y H Shen
- Department of Second Dental Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - F Wang
- Department of Second Dental Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Q G Dai
- Department of Second Dental Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
- Department of Stomatology, Zhang Zhiyuan Academician Work Station, Hainan, Western Central Hospital, Danzhou, Hainan, China
| | - Y Q Wu
- Department of Second Dental Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| |
Collapse
|
6
|
Webbers SD, Aarts CE, Klein B, Koops D, Geissler J, Tool AT, van Bruggen R, van den Akker E, Kuijpers TW. Reduced myeloid commitment and increased uptake by macrophages of stem cell-derived HPS2 neutrophils. Life Sci Alliance 2024; 7:e202302263. [PMID: 38238087 PMCID: PMC10796564 DOI: 10.26508/lsa.202302263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 12/22/2023] [Accepted: 12/27/2023] [Indexed: 01/22/2024] Open
Abstract
Hermansky-Pudlak syndrome type 2 (HPS2) is a rare autosomal recessive disorder, caused by mutations in the AP3B1 gene, encoding the β3A subunit of the adapter protein complex 3. This results in mis-sorting of proteins within the cell. A clinical feature of HPS2 is severe neutropenia. Current HPS2 animal models do not recapitulate the human disease. Hence, we used induced pluripotent stem cells (iPSCs) of an HPS2 patient to study granulopoiesis. Development into CD15POS cells was reduced, but HPS2-derived CD15POS cells differentiated into segmented CD11b+CD16hi neutrophils. These HPS2 neutrophils phenocopied their circulating counterparts showing increased CD63 expression, impaired degranulation capacity, and intact NADPH oxidase activity. Most noticeable was the decrease in neutrophil yield during the final days of HPS2 iPSC cultures. Although neutrophil viability was normal, CD15NEG macrophages were readily phagocytosing neutrophils, contributing to the limited neutrophil output in HPS2. In this iPSC model, HPS2 neutrophil development is affected by a slower rate of development and by macrophage-mediated clearance during neutrophil maturation.
Collapse
Affiliation(s)
- Steven Ds Webbers
- https://ror.org/01fm2fv39 Department of Molecular Hematology, Sanquin Research, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, Netherlands
- Department of Pediatric Immunology, Rheumatology & Infectious Diseases, Emma Children's Hospital, AUMC, University of Amsterdam, Amsterdam, Netherlands
| | - Cathelijn Em Aarts
- https://ror.org/01fm2fv39 Department of Molecular Hematology, Sanquin Research, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, Netherlands
| | - Bart Klein
- https://ror.org/01fm2fv39 Department of Molecular Hematology, Sanquin Research, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, Netherlands
| | - Dané Koops
- https://ror.org/01fm2fv39 Department of Molecular Hematology, Sanquin Research, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, Netherlands
- Department of Pediatric Immunology, Rheumatology & Infectious Diseases, Emma Children's Hospital, AUMC, University of Amsterdam, Amsterdam, Netherlands
| | - Judy Geissler
- https://ror.org/01fm2fv39 Department of Molecular Hematology, Sanquin Research, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, Netherlands
| | - Anton Tj Tool
- https://ror.org/01fm2fv39 Department of Molecular Hematology, Sanquin Research, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, Netherlands
| | - Robin van Bruggen
- https://ror.org/01fm2fv39 Department of Molecular Hematology, Sanquin Research, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, Netherlands
| | - Emile van den Akker
- https://ror.org/01fm2fv39 Department of Hematopoiesis, Sanquin Research Amsterdam, Amsterdam, Netherlands
| | - Taco W Kuijpers
- https://ror.org/01fm2fv39 Department of Molecular Hematology, Sanquin Research, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, Netherlands
- Department of Pediatric Immunology, Rheumatology & Infectious Diseases, Emma Children's Hospital, AUMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
7
|
Signoretto I, Calzetti F, Gasperini S, Bianchetto-Aguilera F, Gardiman E, Finotti G, Tecchio C, Tamassia N, Cassatella MA. Human CD34+/dim neutrophil-committed progenitors do not differentiate into neutrophil-like CXCR1+CD14+CD16- monocytes in vitro. J Leukoc Biol 2024; 115:695-705. [PMID: 38114064 DOI: 10.1093/jleuko/qiad160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/25/2023] [Accepted: 11/28/2023] [Indexed: 12/21/2023] Open
Abstract
The advent of recent cutting-edge technologies has allowed the discovery and characterization of novel progenitors of human neutrophils, including SSCloCD66b+CD15+CD11b-CD49dhiproNeu1s, SSChiCD66b+CD15+CD11b-CD49dintproNeus2s, CD66b+CD15+CD11b+CD49d+CD101-preNeus, and Lin-CD66b+CD117+CD71+eNePs. In this research field, we recently identified CD66b-CD38+CD64dimCD115-, CD34+, and CD34dim/- cells exclusively committed to the neutrophil lineage (which we renamed as CD34+ and CD34dim/- neutrophil-committed progenitors), representing the earliest neutrophil precursors identifiable and sorted by flow cytometry. Moreover, based on their differential CD34 and CD45RA expression, we could identify 4 populations of neutrophil-committed progenitors: CD34+CD45RA-/NCP1s, CD34+CD45RA+/NCP2s, CD34dim/-CD45RA+/NCP3s, and CD34dim/-CD45RA-/NCP4s. This said, a very recent study by Ikeda and coworkers (PMID: 36862552) reported that neutrophil precursors, termed either neutrophil progenitors or "early neutrophil-committed progenitors," would generate immunosuppressive neutrophil-like CXCR1+CD14+CD16- monocytes. Hence, presuming that neutrophil progenitors/"early neutrophil-committed progenitors" correspond to neutrophil-committed progenitors, the selective neutrophil commitment that we attributed to neutrophil-committed progenitors is contradicted by Ikeda and coworkers' article. In this study, by performing a more analytical reevaluation at the phenotypic and molecular levels of the cells generated by neutrophil-committed progenitors 2 and 4 (selected as representatives of neutrophil-committed progenitors), we categorically exclude that neutrophil-committed progenitors generate neutrophil-like CXCR1+CD14+CD16- monocytes. Rather, we provide substantial evidence indicating that the cells generated by neutrophil progenitors/"early neutrophil-committed progenitors" are neutrophilic cells at a different stage of maturation, displaying moderate levels of CD14, instead of neutrophil-like CXCR1+CD14+CD16- monocytes, as pointed by Ikeda and coworkers. Hence, the conclusion that neutrophil progenitors/"early neutrophil-committed progenitors" aberrantly differentiate into neutrophil-like monocytes derives, in our opinion, from data misinterpretation.
Collapse
Affiliation(s)
- Ilaria Signoretto
- Department of Medicine, Section of General Pathology, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy
| | - Federica Calzetti
- Department of Medicine, Section of General Pathology, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy
| | - Sara Gasperini
- Department of Medicine, Section of General Pathology, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy
| | | | - Elisa Gardiman
- Department of Medicine, Section of General Pathology, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy
| | - Giulia Finotti
- Centro Piattaforme Tecnologiche, University of Verona, Piazzale L.A. Scuro 10, 37134 Verona, Italy
| | - Cristina Tecchio
- Department of Medicine, Section of Hematology and Bone Marrow Transplant Unit, University of Verona, Piazzale L.A. Scuro 10, 37134 Verona, Italy
| | - Nicola Tamassia
- Department of Medicine, Section of General Pathology, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy
| | - Marco A Cassatella
- Department of Medicine, Section of General Pathology, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy
| |
Collapse
|
8
|
Fan D, Cong Y, Liu J, Zhang H, Du Z. Spatiotemporal analysis of mRNA-protein relationships enhances transcriptome-based developmental inference. Cell Rep 2024; 43:113928. [PMID: 38461413 DOI: 10.1016/j.celrep.2024.113928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 01/31/2024] [Accepted: 02/22/2024] [Indexed: 03/12/2024] Open
Abstract
Elucidating the complex relationships between mRNA and protein expression at high spatiotemporal resolution is critical for unraveling multilevel gene regulation and enhancing mRNA-based developmental analyses. In this study, we conduct a single-cell analysis of mRNA and protein expression of transcription factors throughout C. elegans embryogenesis. Initially, cellular co-presence of mRNA and protein is low, increasing to a medium-high level (73%) upon factoring in delayed protein synthesis and long-term protein persistence. These factors substantially affect mRNA-protein concordance, leading to potential inaccuracies in mRNA-reliant gene detection and specificity characterization. Building on the learned relationship, we infer protein presence from mRNA expression and demonstrate its utility in identifying tissue-specific genes and elucidating relationships between genes and cells. This approach facilitates identifying the role of sptf-1/SP7 in neuronal lineage development. Collectively, this study provides insights into gene expression dynamics during rapid embryogenesis and approaches for improving the efficacy of transcriptome-based developmental analyses.
Collapse
Affiliation(s)
- Duchangjiang Fan
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Yulin Cong
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Jinyi Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Haoye Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Zhuo Du
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of the Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
9
|
Sollberger G, Brenes AJ, Warner J, Arthur JSC, Howden AJM. Quantitative proteomics reveals tissue-specific, infection-induced and species-specific neutrophil protein signatures. Sci Rep 2024; 14:5966. [PMID: 38472281 PMCID: PMC10933280 DOI: 10.1038/s41598-024-56163-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 03/02/2024] [Indexed: 03/14/2024] Open
Abstract
Neutrophils are one of the first responders to infection and are a key component of the innate immune system through their ability to phagocytose and kill invading pathogens, secrete antimicrobial molecules and produce extracellular traps. Neutrophils are produced in the bone marrow, circulate within the blood and upon immune challenge migrate to the site of infection. We wanted to understand whether this transition shapes the mouse neutrophil protein landscape, how the mouse neutrophil proteome is impacted by systemic infection and perform a comparative analysis of human and mouse neutrophils. Using quantitative mass spectrometry we reveal tissue-specific, infection-induced and species-specific neutrophil protein signatures. We show a high degree of proteomic conservation between mouse bone marrow, blood and peritoneal neutrophils, but also identify key differences in the molecules that these cells express for sensing and responding to their environment. Systemic infection triggers a change in the bone marrow neutrophil population with considerable impact on the core machinery for protein synthesis and DNA replication along with environmental sensors. We also reveal profound differences in mouse and human blood neutrophils, particularly their granule contents. Our proteomics data provides a valuable resource for understanding neutrophil function and phenotypes across species and model systems.
Collapse
Affiliation(s)
- Gabriel Sollberger
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK.
| | - Alejandro J Brenes
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Jordan Warner
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
| | - J Simon C Arthur
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Andrew J M Howden
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK.
| |
Collapse
|
10
|
Co Soriano JC, Tsutsumi S, Ohara D, Hirota K, Kondoh G, Niwa T, Taguchi H, Kadonosono T, Kizaka-Kondoh S. Identification of Surface Markers and Functional Characterization of Myeloid Derived Suppressor Cell-Like Adherent Cells. Adv Biol (Weinh) 2024; 8:e2300159. [PMID: 37986133 DOI: 10.1002/adbi.202300159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 10/11/2023] [Indexed: 11/22/2023]
Abstract
Myeloid-derived suppressor cell (MDSC)-like adherent cells (MLACs) are a recently identified CD11b+ F4/80- myeloid cell subset that can infiltrate tumors early in development and promote their growth. Because of these functions, MLACs play an important role in establishing an immunosuppressive tumor microenvironment (TME). However, the lack of MLAC-specific markers has hampered further characterization of this cell type. This study identifies the gene signature of MLACs by analyzing RNA-sequencing (RNA-seq) and public single-cell RNA-seq data, revealing that MLACs are an independent cell population that are distinct from other intratumoral myeloid cells. After combining proteome analysis of membrane proteins with RNA-seq data, H2-Ab1 and CD11c are indicated as marker proteins that can support the isolation of MLAC subsets from CD11b+ F4/80- myeloid cells by fluorescence-activated cell sorting. The CD11b+ F4/80- H2-Ab1+ and CD11b+ F4/80- CD11c+ MLAC subsets represent approximately half of the MLAC population that is isolated based on their adhesion properties and possess gene signatures and functional properties similar to those of the MLAC population. Additionally, membrane proteome analysis suggests that MLACs express highly heterogeneous surface proteins. This study facilitates an integrated understanding of heterogeneous intratumoral myeloid cells, as well as the molecular and cellular details of the development of an immunosuppressive TME.
Collapse
Affiliation(s)
- John Clyde Co Soriano
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501, Japan
| | - Shiho Tsutsumi
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501, Japan
| | - Daiya Ohara
- Institute for Frontier Life and Medical Sciences, Kyoto University, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Keiji Hirota
- Institute for Frontier Life and Medical Sciences, Kyoto University, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Gen Kondoh
- Institute for Frontier Life and Medical Sciences, Kyoto University, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Tatsuya Niwa
- Institute for Innovative Research, Tokyo Institute of Technology, Yokohama, 226-8503, Japan
| | - Hideki Taguchi
- Institute for Innovative Research, Tokyo Institute of Technology, Yokohama, 226-8503, Japan
| | - Tetsuya Kadonosono
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501, Japan
| | - Shinae Kizaka-Kondoh
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501, Japan
| |
Collapse
|
11
|
Long MB, Howden AJM, Keir HR, Rollings CM, Giam YH, Pembridge T, Delgado L, Abo-Leyah H, Lloyd AF, Sollberger G, Hull R, Gilmour A, Hughes C, New BJM, Cassidy D, Shoemark A, Richardson H, Lamond AI, Cantrell DA, Chalmers JD, Brenes AJ. Extensive acute and sustained changes to neutrophil proteomes post-SARS-CoV-2 infection. Eur Respir J 2024; 63:2300787. [PMID: 38097207 PMCID: PMC10918319 DOI: 10.1183/13993003.00787-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 11/23/2023] [Indexed: 02/15/2024]
Abstract
BACKGROUND Neutrophils are important in the pathophysiology of coronavirus disease 2019 (COVID-19), but the molecular changes contributing to altered neutrophil phenotypes following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection are not fully understood. We used quantitative mass spectrometry-based proteomics to explore neutrophil phenotypes immediately following acute SARS-CoV-2 infection and during recovery. METHODS Prospective observational study of hospitalised patients with PCR-confirmed SARS-CoV-2 infection (May to December 2020). Patients were enrolled within 96 h of admission, with longitudinal sampling up to 29 days. Control groups comprised non-COVID-19 acute lower respiratory tract infection (LRTI) and age-matched noninfected controls. Neutrophils were isolated from peripheral blood and analysed using mass spectrometry. COVID-19 severity and recovery were defined using the World Health Organization ordinal scale. RESULTS Neutrophil proteomes from 84 COVID-19 patients were compared to those from 91 LRTI and 42 control participants. 5800 neutrophil proteins were identified, with >1700 proteins significantly changed in neutrophils from COVID-19 patients compared to noninfected controls. Neutrophils from COVID-19 patients initially all demonstrated a strong interferon signature, but this signature rapidly declined in patients with severe disease. Severe disease was associated with increased abundance of proteins involved in metabolism, immunosuppression and pattern recognition, while delayed recovery from COVID-19 was associated with decreased granule components and reduced abundance of metabolic proteins, chemokine and leukotriene receptors, integrins and inhibitory receptors. CONCLUSIONS SARS-CoV-2 infection results in the sustained presence of circulating neutrophils with distinct proteomes suggesting altered metabolic and immunosuppressive profiles and altered capacities to respond to migratory signals and cues from other immune cells, pathogens or cytokines.
Collapse
Affiliation(s)
- Merete B Long
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
- Indicates equal contribution
| | - Andrew J M Howden
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
- Indicates equal contribution
| | - Holly R Keir
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
- Indicates equal contribution
| | - Christina M Rollings
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
- Indicates equal contribution
| | - Yan Hui Giam
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Thomas Pembridge
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Lilia Delgado
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Hani Abo-Leyah
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Amy F Lloyd
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Gabriel Sollberger
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
- Max Planck Institute for Infection Biology, Berlin, Germany
| | - Rebecca Hull
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Amy Gilmour
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Chloe Hughes
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Benjamin J M New
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Diane Cassidy
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Amelia Shoemark
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Hollian Richardson
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Angus I Lamond
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Doreen A Cantrell
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
| | - James D Chalmers
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
- Indicates joint senior authorship
| | - Alejandro J Brenes
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, UK
- Indicates joint senior authorship
| |
Collapse
|
12
|
Danne C, Skerniskyte J, Marteyn B, Sokol H. Neutrophils: from IBD to the gut microbiota. Nat Rev Gastroenterol Hepatol 2024; 21:184-197. [PMID: 38110547 DOI: 10.1038/s41575-023-00871-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/10/2023] [Indexed: 12/20/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory condition of the gastrointestinal tract that results from dysfunction in innate and/or adaptive immune responses. Impaired innate immunity, which leads to lack of control of an altered intestinal microbiota and to activation of the adaptive immune system, promotes a secondary inflammatory response that is responsible for tissue damage. Neutrophils are key players in innate immunity in IBD, but their roles have been neglected compared with those of other immune cells. The latest studies on neutrophils in IBD have revealed unexpected complexities, with heterogeneous populations and dual functions, both deleterious and protective, for the host. In parallel, interconnections between disease development, intestinal microbiota and neutrophils have been highlighted. Numerous IBD susceptibility genes (such as NOD2, NCF4, LRRK2, CARD9) are involved in neutrophil functions related to defence against microorganisms. Moreover, severe monogenic diseases involving dysfunctional neutrophils, including chronic granulomatous disease, are characterized by intestinal inflammation that mimics IBD and by alterations in the intestinal microbiota. This observation demonstrates the dialogue between neutrophils, gut inflammation and the microbiota. Neutrophils affect microbiota composition and function in several ways. In return, microbial factors, including metabolites, regulate neutrophil production and function directly and indirectly. It is crucial to further investigate the diverse roles played by neutrophils in host-microbiota interactions, both at steady state and in inflammatory conditions, to develop new IBD therapies. In this Review, we discuss the roles of neutrophils in IBD, in light of emerging evidence proving strong interconnections between neutrophils and the gut microbiota, especially in an inflammatory context.
Collapse
Affiliation(s)
- Camille Danne
- Sorbonne Université, INSERM UMRS-938, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint-Antoine, Service de Gastroentérologie, Paris, France.
- Paris Center For Microbiome Medicine (PaCeMM) FHU, Paris, France.
| | - Jurate Skerniskyte
- CNRS, UPR 9002, Université de Strasbourg, Institut de Biologie Moléculaire et Cellulaire, Architecture et Réactivité de l'ARN, Strasbourg, France
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Benoit Marteyn
- CNRS, UPR 9002, Université de Strasbourg, Institut de Biologie Moléculaire et Cellulaire, Architecture et Réactivité de l'ARN, Strasbourg, France
- University of Strasbourg Institute for Advanced Study (USIAS), Strasbourg, France
- Institut Pasteur, Université de Paris, Inserm 1225 Unité de Pathogenèse des Infections Vasculaires, Paris, France
| | - Harry Sokol
- Sorbonne Université, INSERM UMRS-938, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint-Antoine, Service de Gastroentérologie, Paris, France
- Paris Center For Microbiome Medicine (PaCeMM) FHU, Paris, France
- Université Paris-Saclay, INRAe, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| |
Collapse
|
13
|
Thind MK, Uhlig HH, Glogauer M, Palaniyar N, Bourdon C, Gwela A, Lancioni CL, Berkley JA, Bandsma RHJ, Farooqui A. A metabolic perspective of the neutrophil life cycle: new avenues in immunometabolism. Front Immunol 2024; 14:1334205. [PMID: 38259490 PMCID: PMC10800387 DOI: 10.3389/fimmu.2023.1334205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 12/15/2023] [Indexed: 01/24/2024] Open
Abstract
Neutrophils are the most abundant innate immune cells. Multiple mechanisms allow them to engage a wide range of metabolic pathways for biosynthesis and bioenergetics for mediating biological processes such as development in the bone marrow and antimicrobial activity such as ROS production and NET formation, inflammation and tissue repair. We first discuss recent work on neutrophil development and functions and the metabolic processes to regulate granulopoiesis, neutrophil migration and trafficking as well as effector functions. We then discuss metabolic syndromes with impaired neutrophil functions that are influenced by genetic and environmental factors of nutrient availability and usage. Here, we particularly focus on the role of specific macronutrients, such as glucose, fatty acids, and protein, as well as micronutrients such as vitamin B3, in regulating neutrophil biology and how this regulation impacts host health. A special section of this review primarily discusses that the ways nutrient deficiencies could impact neutrophil biology and increase infection susceptibility. We emphasize biochemical approaches to explore neutrophil metabolism in relation to development and functions. Lastly, we discuss opportunities and challenges to neutrophil-centered therapeutic approaches in immune-driven diseases and highlight unanswered questions to guide future discoveries.
Collapse
Affiliation(s)
- Mehakpreet K Thind
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
- The Childhood Acute Illness & Nutrition Network (CHAIN), Nairobi, Kenya
| | - Holm H Uhlig
- Translational Gastroenterology Unit, Experimental Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Michael Glogauer
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
- Department of Dental Oncology and Maxillofacial Prosthetics, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Nades Palaniyar
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
- Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Celine Bourdon
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
- The Childhood Acute Illness & Nutrition Network (CHAIN), Nairobi, Kenya
| | - Agnes Gwela
- The Childhood Acute Illness & Nutrition Network (CHAIN), Nairobi, Kenya
- Kenya Medical Research Institute (KEMRI)/Wellcome Trust Research Programme, Centre for Geographic Medicine Research, Kilifi, Kenya
| | - Christina L Lancioni
- The Childhood Acute Illness & Nutrition Network (CHAIN), Nairobi, Kenya
- Department of Pediatrics, Oregon Health and Science University, Portland, OR, United States
| | - James A Berkley
- The Childhood Acute Illness & Nutrition Network (CHAIN), Nairobi, Kenya
- Kenya Medical Research Institute (KEMRI)/Wellcome Trust Research Programme, Centre for Geographic Medicine Research, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, United Kingdom
| | - Robert H J Bandsma
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
- The Childhood Acute Illness & Nutrition Network (CHAIN), Nairobi, Kenya
- Laboratory of Pediatrics, Center for Liver, Digestive, and Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
- Division of Gastroenterology, Hepatology, and Nutrition, The Hospital for Sick Children, Toronto, ON, Canada
| | - Amber Farooqui
- Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
- The Childhood Acute Illness & Nutrition Network (CHAIN), Nairobi, Kenya
- Omega Laboratories Inc, Mississauga, ON, Canada
| |
Collapse
|
14
|
Vanhaver C, Aboubakar Nana F, Delhez N, Luyckx M, Hirsch T, Bayard A, Houbion C, Dauguet N, Brochier A, van der Bruggen P, Bruger AM. Immunosuppressive low-density neutrophils in the blood of cancer patients display a mature phenotype. Life Sci Alliance 2024; 7:e202302332. [PMID: 37931958 PMCID: PMC10628041 DOI: 10.26508/lsa.202302332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 11/08/2023] Open
Abstract
The presence of human neutrophils in the tumor microenvironment is strongly correlated to poor overall survival. Most previous studies have focused on the immunosuppressive capacities of low-density neutrophils (LDN), also referred to as granulocytic myeloid-derived suppressor cells, which are elevated in number in the blood of many cancer patients. We observed two types of LDN in the blood of lung cancer and ovarian carcinoma patients: CD45high LDN, which suppressed T-cell proliferation and displayed mature morphology, and CD45low LDN, which were immature and non-suppressive. We simultaneously evaluated the classical normal-density neutrophils (NDN) and, when available, tumor-associated neutrophils. We observed that NDN from cancer patients suppressed T-cell proliferation, and NDN from healthy donors did not, despite few transcriptomic differences. Hence, the immunosuppression mediated by neutrophils in the blood of cancer patients is not dependent on the cells' density but rather on their maturity.
Collapse
Affiliation(s)
- Christophe Vanhaver
- https://ror.org/022em3k58 Institut de Duve, Université Catholique de Louvain, Brussels, Belgium
| | - Frank Aboubakar Nana
- https://ror.org/022em3k58 Institut de Duve, Université Catholique de Louvain, Brussels, Belgium
- Service de Pneumologie, Cliniques Universitaires Saint-Luc, Brussels, Belgium
- Institut de Recherche Expérimentale et Clinique (IREC)/Pôle de Pneumologie, Université Catholique de Louvain, Brussels, Belgium
| | - Nicolas Delhez
- https://ror.org/022em3k58 Institut de Duve, Université Catholique de Louvain, Brussels, Belgium
| | - Mathieu Luyckx
- https://ror.org/022em3k58 Institut de Duve, Université Catholique de Louvain, Brussels, Belgium
- Service de Gynécologie et Andrologie, Cliniques Universitaires Saint-Luc, Brussels, Belgium
- Centre de Chirurgie Oncologique, Institut Roi Albert II, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Thibault Hirsch
- https://ror.org/022em3k58 Institut de Duve, Université Catholique de Louvain, Brussels, Belgium
| | - Alexandre Bayard
- https://ror.org/022em3k58 Institut de Duve, Université Catholique de Louvain, Brussels, Belgium
| | - Camille Houbion
- https://ror.org/022em3k58 Institut de Duve, Université Catholique de Louvain, Brussels, Belgium
| | - Nicolas Dauguet
- https://ror.org/022em3k58 Institut de Duve, Université Catholique de Louvain, Brussels, Belgium
| | - Alice Brochier
- Hematology Department of Laboratory Medicine, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Pierre van der Bruggen
- https://ror.org/022em3k58 Institut de Duve, Université Catholique de Louvain, Brussels, Belgium
| | - Annika M Bruger
- https://ror.org/022em3k58 Institut de Duve, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
15
|
Zheng L, Rang M, Fuchs C, Keß A, Wunsch M, Hentschel J, Hsiao CC, Kleber C, Osterhoff G, Aust G. The Posttraumatic Increase of the Adhesion GPCR EMR2/ ADGRE2 on Circulating Neutrophils Is Not Related to Injury Severity. Cells 2023; 12:2657. [PMID: 37998392 PMCID: PMC10670733 DOI: 10.3390/cells12222657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/05/2023] [Accepted: 11/16/2023] [Indexed: 11/25/2023] Open
Abstract
Trauma triggers a rapid innate immune response to aid the clearance of damaged/necrotic cells and their released damage-associated molecular pattern (DAMP). Here, we monitored the expression of EMR2/ADGRE2, involved in the functional regulation of innate immune cells, on circulating neutrophils in very severely and moderately/severely injured patients up to 240 h after trauma. Notably, neutrophilic EMR2 showed a uniform, injury severity- and type of injury-independent posttraumatic course in all patients. The percentage of EMR2+ neutrophils and their EMR2 level increased and peaked 48 h after trauma. Afterwards, they declined and normalized in some, but not all, patients. Circulating EMR2+ compared to EMR2- neutrophils express less CD62L and more CD11c, a sign of activation. Neutrophilic EMR2 regulation was verified in vitro. Remarkably, it increased, depending on extracellular calcium, in controls as well. Cytokines, enhanced in patients immediately after trauma, and sera of patients did not further affect this neutrophilic EMR2 increase, whereas apoptosis induction disrupted it. Likely the damaged/necrotic cells/DAMPs, unavoidable during neutrophil culture, stimulate the neutrophilic EMR2 increase. In summary, the rapidly increased absolute number of neutrophils, especially present in very severely injured patients, together with upregulated neutrophilic EMR2, may expand our in vivo capacity to react to and finally clear damaged/necrotic cells/DAMPs after trauma.
Collapse
Affiliation(s)
- Leyu Zheng
- Research Laboratories and Department of Orthopaedics, Trauma and Plastic Surgery (OUP), Leipzig University and University Hospital Leipzig, 04103 Leipzig, Germany; (L.Z.); (M.R.); (C.F.); (A.K.); (M.W.); (C.K.); (G.O.)
| | - Moujie Rang
- Research Laboratories and Department of Orthopaedics, Trauma and Plastic Surgery (OUP), Leipzig University and University Hospital Leipzig, 04103 Leipzig, Germany; (L.Z.); (M.R.); (C.F.); (A.K.); (M.W.); (C.K.); (G.O.)
| | - Carolin Fuchs
- Research Laboratories and Department of Orthopaedics, Trauma and Plastic Surgery (OUP), Leipzig University and University Hospital Leipzig, 04103 Leipzig, Germany; (L.Z.); (M.R.); (C.F.); (A.K.); (M.W.); (C.K.); (G.O.)
| | - Annette Keß
- Research Laboratories and Department of Orthopaedics, Trauma and Plastic Surgery (OUP), Leipzig University and University Hospital Leipzig, 04103 Leipzig, Germany; (L.Z.); (M.R.); (C.F.); (A.K.); (M.W.); (C.K.); (G.O.)
| | - Mandy Wunsch
- Research Laboratories and Department of Orthopaedics, Trauma and Plastic Surgery (OUP), Leipzig University and University Hospital Leipzig, 04103 Leipzig, Germany; (L.Z.); (M.R.); (C.F.); (A.K.); (M.W.); (C.K.); (G.O.)
| | - Julia Hentschel
- Institute of Human Genetics, Leipzig University and University Hospital Leipzig, 04103 Leipzig, Germany;
| | - Cheng-Chih Hsiao
- Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, 1105 AZ Amsterdam, The Netherlands;
| | - Christian Kleber
- Research Laboratories and Department of Orthopaedics, Trauma and Plastic Surgery (OUP), Leipzig University and University Hospital Leipzig, 04103 Leipzig, Germany; (L.Z.); (M.R.); (C.F.); (A.K.); (M.W.); (C.K.); (G.O.)
| | - Georg Osterhoff
- Research Laboratories and Department of Orthopaedics, Trauma and Plastic Surgery (OUP), Leipzig University and University Hospital Leipzig, 04103 Leipzig, Germany; (L.Z.); (M.R.); (C.F.); (A.K.); (M.W.); (C.K.); (G.O.)
| | - Gabriela Aust
- Research Laboratories and Department of Orthopaedics, Trauma and Plastic Surgery (OUP), Leipzig University and University Hospital Leipzig, 04103 Leipzig, Germany; (L.Z.); (M.R.); (C.F.); (A.K.); (M.W.); (C.K.); (G.O.)
- Research Laboratories and Department of Visceral, Transplantation, Vascular and Thoracic Surgery (VTTG), Leipzig University and University Hospital Leipzig, 04103 Leipzig, Germany
| |
Collapse
|
16
|
Sitarska E, Almeida SD, Beckwith MS, Stopp J, Czuchnowski J, Siggel M, Roessner R, Tschanz A, Ejsing C, Schwab Y, Kosinski J, Sixt M, Kreshuk A, Erzberger A, Diz-Muñoz A. Sensing their plasma membrane curvature allows migrating cells to circumvent obstacles. Nat Commun 2023; 14:5644. [PMID: 37704612 PMCID: PMC10499897 DOI: 10.1038/s41467-023-41173-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 08/22/2023] [Indexed: 09/15/2023] Open
Abstract
To navigate through diverse tissues, migrating cells must balance persistent self-propelled motion with adaptive behaviors to circumvent obstacles. We identify a curvature-sensing mechanism underlying obstacle evasion in immune-like cells. Specifically, we propose that actin polymerization at the advancing edge of migrating cells is inhibited by the curvature-sensitive BAR domain protein Snx33 in regions with inward plasma membrane curvature. The genetic perturbation of this machinery reduces the cells' capacity to evade obstructions combined with faster and more persistent cell migration in obstacle-free environments. Our results show how cells can read out their surface topography and utilize actin and plasma membrane biophysics to interpret their environment, allowing them to adaptively decide if they should move ahead or turn away. On the basis of our findings, we propose that the natural diversity of BAR domain proteins may allow cells to tune their curvature sensing machinery to match the shape characteristics in their environment.
Collapse
Affiliation(s)
- Ewa Sitarska
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117, Heidelberg, Germany
- Collaboration for joint PhD degree between EMBL and Heidelberg University, Faculty of Biosciences, EMBL and Heidelberg University, Heidelberg, Germany
| | - Silvia Dias Almeida
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117, Heidelberg, Germany
- Division of Medical Image Computing, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | | | - Julian Stopp
- Institute of Science and Technology Austria, 3400, Klosterneuburg, Austria
| | - Jakub Czuchnowski
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117, Heidelberg, Germany
| | - Marc Siggel
- EMBL Hamburg, European Molecular Biology Laboratory, 22607, Hamburg, Germany
- Centre for Structural Systems Biology, 22607, Hamburg, Germany
| | - Rita Roessner
- EMBL Hamburg, European Molecular Biology Laboratory, 22607, Hamburg, Germany
- Centre for Structural Systems Biology, 22607, Hamburg, Germany
| | - Aline Tschanz
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117, Heidelberg, Germany
- Collaboration for joint PhD degree between EMBL and Heidelberg University, Faculty of Biosciences, EMBL and Heidelberg University, Heidelberg, Germany
| | - Christer Ejsing
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117, Heidelberg, Germany
- Department of Biochemistry and Molecular Biology, Villum Center for Bioanalytical Sciences, University of Southern Denmark, Campusvej 55, 5230, Odense, Denmark
| | - Yannick Schwab
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117, Heidelberg, Germany
| | - Jan Kosinski
- EMBL Hamburg, European Molecular Biology Laboratory, 22607, Hamburg, Germany
- Centre for Structural Systems Biology, 22607, Hamburg, Germany
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, 69117, Heidelberg, Germany
| | - Michael Sixt
- Institute of Science and Technology Austria, 3400, Klosterneuburg, Austria
| | - Anna Kreshuk
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117, Heidelberg, Germany
| | - Anna Erzberger
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117, Heidelberg, Germany
| | - Alba Diz-Muñoz
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117, Heidelberg, Germany.
| |
Collapse
|
17
|
Metzemaekers M, Malengier-Devlies B, Gouwy M, De Somer L, Cunha FDQ, Opdenakker G, Proost P. Fast and furious: The neutrophil and its armamentarium in health and disease. Med Res Rev 2023; 43:1537-1606. [PMID: 37036061 DOI: 10.1002/med.21958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 12/27/2022] [Accepted: 03/24/2023] [Indexed: 04/11/2023]
Abstract
Neutrophils are powerful effector cells leading the first wave of acute host-protective responses. These innate leukocytes are endowed with oxidative and nonoxidative defence mechanisms, and play well-established roles in fighting invading pathogens. With microbicidal weaponry largely devoid of specificity and an all-too-well recognized toxicity potential, collateral damage may occur in neutrophil-rich diseases. However, emerging evidence suggests that neutrophils are more versatile, heterogeneous, and sophisticated cells than initially thought. At the crossroads of innate and adaptive immunity, neutrophils demonstrate their multifaceted functions in infectious and noninfectious pathologies including cancer, autoinflammation, and autoimmune diseases. Here, we discuss the kinetics of neutrophils and their products of activation from bench to bedside during health and disease, and provide an overview of the versatile functions of neutrophils as key modulators of immune responses and physiological processes. We focus specifically on those activities and concepts that have been validated with primary human cells.
Collapse
Affiliation(s)
- Mieke Metzemaekers
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Bert Malengier-Devlies
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Mieke Gouwy
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Lien De Somer
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
- Division of Pediatric Rheumatology, University Hospital Leuven, Leuven, Belgium
- European Reference Network for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (RITA) at the University Hospital Leuven, Leuven, Belgium
| | | | - Ghislain Opdenakker
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
18
|
Carbonell AU, Freire-Cobo C, Deyneko IV, Dobariya S, Erdjument-Bromage H, Clipperton-Allen AE, Page DT, Neubert TA, Jordan BA. Comparing synaptic proteomes across five mouse models for autism reveals converging molecular similarities including deficits in oxidative phosphorylation and Rho GTPase signaling. Front Aging Neurosci 2023; 15:1152562. [PMID: 37255534 PMCID: PMC10225639 DOI: 10.3389/fnagi.2023.1152562] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 04/17/2023] [Indexed: 06/01/2023] Open
Abstract
Specific and effective treatments for autism spectrum disorder (ASD) are lacking due to a poor understanding of disease mechanisms. Here we test the idea that similarities between diverse ASD mouse models are caused by deficits in common molecular pathways at neuronal synapses. To do this, we leverage the availability of multiple genetic models of ASD that exhibit shared synaptic and behavioral deficits and use quantitative mass spectrometry with isobaric tandem mass tagging (TMT) to compare their hippocampal synaptic proteomes. Comparative analyses of mouse models for Fragile X syndrome (Fmr1 knockout), cortical dysplasia focal epilepsy syndrome (Cntnap2 knockout), PTEN hamartoma tumor syndrome (Pten haploinsufficiency), ANKS1B syndrome (Anks1b haploinsufficiency), and idiopathic autism (BTBR+) revealed several common altered cellular and molecular pathways at the synapse, including changes in oxidative phosphorylation, and Rho family small GTPase signaling. Functional validation of one of these aberrant pathways, Rac1 signaling, confirms that the ANKS1B model displays altered Rac1 activity counter to that observed in other models, as predicted by the bioinformatic analyses. Overall similarity analyses reveal clusters of synaptic profiles, which may form the basis for molecular subtypes that explain genetic heterogeneity in ASD despite a common clinical diagnosis. Our results suggest that ASD-linked susceptibility genes ultimately converge on common signaling pathways regulating synaptic function and propose that these points of convergence are key to understanding the pathogenesis of this disorder.
Collapse
Affiliation(s)
- Abigail U. Carbonell
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Carmen Freire-Cobo
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Ilana V. Deyneko
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Saunil Dobariya
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Hediye Erdjument-Bromage
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, United States
| | - Amy E. Clipperton-Allen
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL, United States
| | - Damon T. Page
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL, United States
| | - Thomas A. Neubert
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, United States
| | - Bryen A. Jordan
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, United States
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
19
|
Martinez-Sanz P, Laurent ARG, Slot E, Hoogenboezem M, Bąbała N, van Bruggen R, Rongvaux A, Flavell RA, Tytgat GAM, Franke K, Matlung HL, Kuijpers TW, Amsen D, Karrich JJ. Humanized MISTRG as a preclinical in vivo model to study human neutrophil-mediated immune processes. Front Immunol 2023; 14:1105103. [PMID: 36969261 PMCID: PMC10032520 DOI: 10.3389/fimmu.2023.1105103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 02/22/2023] [Indexed: 03/11/2023] Open
Abstract
IntroductionMISTRG mice have been genetically modified to allow development of a human myeloid compartment from engrafted human CD34+ haemopoietic stem cells, making them particularly suited to study the human innate immune system in vivo. Here, we characterized the human neutrophil population in these mice to establish a model that can be used to study the biology and contribution in immune processes of these cells in vivo.Methods and resultsWe could isolate human bone marrow neutrophils from humanized MISTRG mice and confirmed that all neutrophil maturation stages from promyelocytes (CD11b–CD16–) to end-stage segmented cells (CD11b+CD16+) were present. We documented that these cells possessed normal functional properties, including degranulation, reactive oxygen species production, adhesion, and antibody-dependent cellular cytotoxicity towards antibody-opsonized tumor cells ex vivo. The acquisition of functional capacities positively correlated with the maturation state of the cell. We found that human neutrophils were retained in the bone marrow of humanized MISTRG mice during steady state. However, the mature segmented CD11b+CD16+ human neutrophils were released from the bone marrow in response to two well-established neutrophil-mobilizing agents (i.e., G-CSF and/or CXCR4 antagonist Plerixafor). Moreover, the neutrophil population in the humanized MISTRG mice actively reacted to thioglycolate-induced peritonitis and could infiltrate implanted human tumors, as shown by flow cytometry and fluorescent microscopy.DiscussionThese results show that functional human neutrophils are generated and can be studied in vivo using the humanized MISTRG mice, providing a model to study the various functions of neutrophils in inflammation and in tumors.
Collapse
Affiliation(s)
- Paula Martinez-Sanz
- Sanquin Research and Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- *Correspondence: Paula Martinez-Sanz, ; Julien J. Karrich, ; Derk Amsen,
| | - Adrien R. G. Laurent
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Edith Slot
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Mark Hoogenboezem
- Sanquin Research and Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Nikolina Bąbała
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Robin van Bruggen
- Sanquin Research and Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Anthony Rongvaux
- Department of Immunology, University of Washington, Seattle, WA, United States
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, WA, United States
| | - Richard A. Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, United States
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, United States
| | - Godelieve A. M. Tytgat
- Princess Maxima Center for Pediatric Oncology, Department of Pediatric Oncology, Utrecht, Netherlands
| | - Katka Franke
- Sanquin Research and Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Hanke L. Matlung
- Sanquin Research and Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Taco W. Kuijpers
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Rheumatology and Infectious Diseases, Emma Children's Hospital, Department of Pediatric Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Derk Amsen
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- *Correspondence: Paula Martinez-Sanz, ; Julien J. Karrich, ; Derk Amsen,
| | - Julien J. Karrich
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- *Correspondence: Paula Martinez-Sanz, ; Julien J. Karrich, ; Derk Amsen,
| |
Collapse
|
20
|
Segal BH, Giridharan T, Suzuki S, Khan ANH, Zsiros E, Emmons TR, Yaffe MB, Gankema AAF, Hoogeboom M, Goetschalckx I, Matlung HL, Kuijpers TW. Neutrophil interactions with T cells, platelets, endothelial cells, and of course tumor cells. Immunol Rev 2023; 314:13-35. [PMID: 36527200 PMCID: PMC10174640 DOI: 10.1111/imr.13178] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Neutrophils sense microbes and host inflammatory mediators, and traffic to sites of infection where they direct a broad armamentarium of antimicrobial products against pathogens. Neutrophils are also activated by damage-associated molecular patterns (DAMPs), which are products of cellular injury that stimulate the innate immune system through pathways that are similar to those activated by microbes. Neutrophils and platelets become activated by injury, and cluster and cross-signal to each other with the cumulative effect of driving antimicrobial defense and hemostasis. In addition, neutrophil extracellular traps are extracellular chromatin and granular constituents that are generated in response to microbial and damage motifs and are pro-thrombotic and injurious. Although neutrophils can worsen tissue injury, neutrophils may also have a role in facilitating wound repair following injury. A central theme of this review relates to how critical functions of neutrophils that evolved to respond to infection and damage modulate the tumor microenvironment (TME) in ways that can promote or limit tumor progression. Neutrophils are reprogrammed by the TME, and, in turn, can cross-signal to tumor cells and reshape the immune landscape of tumors. Importantly, promising new therapeutic strategies have been developed to target neutrophil recruitment and function to make cancer immunotherapy more effective.
Collapse
Affiliation(s)
- Brahm H Segal
- Department of Internal Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
- Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Thejaswini Giridharan
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Sora Suzuki
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Anm Nazmul H Khan
- Department of Internal Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Emese Zsiros
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Tiffany R Emmons
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Michael B Yaffe
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Angela A F Gankema
- Department of Molecular Hematology, Sanquin Research, University of Amsterdam, Amsterdam, The Netherlands
| | - Mark Hoogeboom
- Department of Molecular Hematology, Sanquin Research, University of Amsterdam, Amsterdam, The Netherlands
| | - Ines Goetschalckx
- Department of Molecular Hematology, Sanquin Research, University of Amsterdam, Amsterdam, The Netherlands
| | - Hanke L Matlung
- Department of Molecular Hematology, Sanquin Research, University of Amsterdam, Amsterdam, The Netherlands
| | - Taco W Kuijpers
- Department of Molecular Hematology, Sanquin Research, University of Amsterdam, Amsterdam, The Netherlands
- Department of Pediatric Immunology, Rheumatology and Infectious Disease, Emma Children's Hospital Amsterdam University Medical Center (Amsterdam UMC), University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
21
|
Danan CH, Katada K, Parham LR, Hamilton KE. Spatial transcriptomics add a new dimension to our understanding of the gut. Am J Physiol Gastrointest Liver Physiol 2023; 324:G91-G98. [PMID: 36472345 PMCID: PMC9870576 DOI: 10.1152/ajpgi.00191.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 01/19/2023]
Abstract
The profound complexity of the intestinal mucosa demands a spatial approach to the study of gut transcriptomics. Although single-cell RNA sequencing has revolutionized our ability to survey the diverse cell types of the intestine, knowledge of cell type alone cannot fully describe the cells that make up the intestinal mucosa. During homeostasis and disease, dramatic gradients of oxygen, nutrients, extracellular matrix proteins, morphogens, and microbiota collectively dictate intestinal cell state, and only spatial techniques can articulate differences in cellular transcriptomes at this level. Spatial transcriptomic techniques assign transcriptomic data to precise regions in a tissue of interest. In recent years, these protocols have become increasingly accessible, and their application in the intestinal mucosa has exploded in popularity. In the gut, spatial transcriptomics typically involve the application of tissue sections to spatially barcoded RNA sequencing or laser capture microdissection followed by RNA sequencing. In combination with single-cell RNA sequencing, these spatial sequencing approaches allow for the construction of spatial transcriptional maps at pseudosingle-cell resolution. In this review, we describe the spatial transcriptomic technologies recently applied in the gut and the previously unattainable discoveries that they have produced.
Collapse
Affiliation(s)
- Charles H Danan
- Medical Scientist Training Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Kay Katada
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Louis R Parham
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Kathryn E Hamilton
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
22
|
Harbig TA, Fratte J, Krone M, Nieselt K. OmicsTIDE: interactive exploration of trends in multi-omics data. BIOINFORMATICS ADVANCES 2023; 3:vbac093. [PMID: 36698763 PMCID: PMC9869718 DOI: 10.1093/bioadv/vbac093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/18/2022] [Accepted: 12/06/2022] [Indexed: 01/22/2023]
Abstract
Motivation The increasing amount of data produced by omics technologies has enabled researchers to study phenomena across multiple omics layers. Besides data-driven analysis strategies, interactive visualization tools have been developed for a more transparent analysis. However, most state-of-the-art tools do not reconstruct the impact of a single omics layer on the integration result. Results We developed a data classification scheme focusing on different aspects of multi-omics datasets for a systemic understanding. Based on this classification, we developed the Omics Trend-comparing Interactive Data Explorer (OmicsTIDE), an interactive visualization tool for the comparison of gene-based quantitative omics data. The tool consists of a computational part that clusters omics datasets to determine trends and an interactive visualization. The trends are visualized as profile plots and are connected by a Sankey diagram that allows for an interactive pairwise trend comparison to discover concordant and discordant trends. Moreover, large-scale omics datasets are broken down into small subsets that can be analyzed functionally using Gene Ontology enrichment within few analysis steps. We demonstrate the interactive analysis using OmicsTIDE with two case studies focusing on different experimental designs. Availability and implementation OmicsTIDE is a web tool available via http://omicstide-tuevis.cs.uni-tuebingen.de/. Supplementary information Supplementary data are available at Bioinformatics Advances online.
Collapse
Affiliation(s)
- Theresa A Harbig
- Institute for Bioinformatics and Medical Informatics, University of Tuebingen, Tuebingen 72076, Germany
| | - Julian Fratte
- Institute for Bioinformatics and Medical Informatics, University of Tuebingen, Tuebingen 72076, Germany
| | - Michael Krone
- Institute for Bioinformatics and Medical Informatics, University of Tuebingen, Tuebingen 72076, Germany
| | - Kay Nieselt
- Institute for Bioinformatics and Medical Informatics, University of Tuebingen, Tuebingen 72076, Germany
| |
Collapse
|
23
|
Gomes F, Alfson K, Junqueira M. Editorial: The application of OMICS technologies to interrogate host-virus interactions. Front Cell Infect Microbiol 2022; 12:1050012. [DOI: 10.3389/fcimb.2022.1050012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 10/21/2022] [Indexed: 11/16/2022] Open
|
24
|
Talal S, Mona K, Karem A, Yaniv L, Reut HM, Ariel S, Moran AK, Harel E, Campisi-Pinto S, Mahmoud AA, Raul C, David T, Gil BS, Idan C. Neutrophil degranulation and severely impaired extracellular trap formation at the basis of susceptibility to infections of hemodialysis patients. BMC Med 2022; 20:364. [PMID: 36284314 PMCID: PMC9597999 DOI: 10.1186/s12916-022-02564-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 09/12/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Chronic kidney disease patients are at increased risk of mortality with cardiovascular diseases and infections as the two leading causes of death for end-stage kidney disease treated with hemodialysis (HD). Mortality from bacterial infections in HD patients is estimated to be 100-1000 times higher than in the healthy population. METHODS We comprehensively characterized highly pure circulating neutrophils from HD and healthy donors. RESULTS Protein levels and transcriptome of HD patients' neutrophils indicated massive neutrophil degranulation with a dramatic reduction in reactive oxygen species (ROS) production during an oxidative burst and defective oxidative cellular signaling. Moreover, HD neutrophils exhibit severely impaired ability to generate extracellular NET formation (NETosis) in NADPH oxidase-dependent or independent pathways, reflecting their loss of capacity to kill extracellular bacteria. Ectopic hydrogen peroxidase (H2O2) or recombinant human SOD-1 (rSOD-1) partly restores and improves the extent of HD dysfunctional neutrophil NET formation. CONCLUSIONS Our report is one of the first singular examples of severe and chronic impairment of NET formation leading to substantial clinical susceptibility to bacteremia that most likely results from the metabolic and environmental milieu typical to HD patients and not by common human genetic deficiencies. In this manner, aberrant gene expression and differential exocytosis of distinct granule populations could reflect the chronic defect in neutrophil functionality and their diminished ability to induce NETosis. Therefore, our findings suggest that targeting NETosis in HD patients may reduce infections, minimize their severity, and decrease the mortality rate from infections in this patient population.
Collapse
Affiliation(s)
- Salti Talal
- Oncology & Hematology Division, Cancer Center, Emek Medical Center, 21 Yitzhak Rabin Blvd, 1834111, Afula, Israel
- Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 320002, Haifa, Israel
| | - Khoury Mona
- Oncology & Hematology Division, Cancer Center, Emek Medical Center, 21 Yitzhak Rabin Blvd, 1834111, Afula, Israel
- Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 320002, Haifa, Israel
| | - Awad Karem
- Nephrology Department, Emek Medical Center, Afula, Israel
| | | | | | - Shemesh Ariel
- Biomedical Core Facility, Bruce Rappaport Faculty of Medicine Technion-Israel, Haifa, Israel
| | | | - Eitam Harel
- Emek Medical Center, Clinical Laboratories, Clalit, Afula, Israel
| | | | - Abu-Amna Mahmoud
- Oncology & Hematology Division, Cancer Center, Emek Medical Center, 21 Yitzhak Rabin Blvd, 1834111, Afula, Israel
| | - Colodner Raul
- Emek Medical Center, Clinical Laboratories, Clalit, Afula, Israel
| | - Tovbin David
- Nephrology Department, Emek Medical Center, Afula, Israel
| | - Bar-Sela Gil
- Oncology & Hematology Division, Cancer Center, Emek Medical Center, 21 Yitzhak Rabin Blvd, 1834111, Afula, Israel.
- Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 320002, Haifa, Israel.
| | - Cohen Idan
- Oncology & Hematology Division, Cancer Center, Emek Medical Center, 21 Yitzhak Rabin Blvd, 1834111, Afula, Israel.
| |
Collapse
|
25
|
Bonadonna M, Altamura S, Tybl E, Palais G, Qatato M, Polycarpou-Schwarz M, Schneider M, Kalk C, Rüdiger W, Ertl A, Anstee N, Bogeska R, Helm D, Milsom MD, Galy B. Iron regulatory protein (IRP)-mediated iron homeostasis is critical for neutrophil development and differentiation in the bone marrow. SCIENCE ADVANCES 2022; 8:eabq4469. [PMID: 36197975 PMCID: PMC9534496 DOI: 10.1126/sciadv.abq4469] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 08/18/2022] [Indexed: 06/01/2023]
Abstract
Iron is mostly devoted to the hemoglobinization of erythrocytes for oxygen transport. However, emerging evidence points to a broader role for the metal in hematopoiesis, including the formation of the immune system. Iron availability in mammalian cells is controlled by iron-regulatory protein 1 (IRP1) and IRP2. We report that global disruption of both IRP1 and IRP2 in adult mice impairs neutrophil development and differentiation in the bone marrow, yielding immature neutrophils with abnormally high glycolytic and autophagic activity, resulting in neutropenia. IRPs promote neutrophil differentiation in a cell intrinsic manner by securing cellular iron supply together with transcriptional control of neutropoiesis to facilitate differentiation to fully mature neutrophils. Unlike neutrophils, monocyte count was not affected by IRP and iron deficiency, suggesting a lineage-specific effect of iron on myeloid output. This study unveils the previously unrecognized importance of IRPs and iron metabolism in the formation of a major branch of the innate immune system.
Collapse
Affiliation(s)
- Michael Bonadonna
- German Cancer Research Center, “Division of Virus-Associated Carcinogenesis”, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Biosciences Faculty, University of Heidelberg, 69120 Heidelberg, Germany
| | - Sandro Altamura
- University of Heidelberg, Department of Pediatric Hematology, Oncology and Immunology, Im Neuenheimer Feld 350, 69120 Heidelberg, Germany
| | - Elisabeth Tybl
- German Cancer Research Center, “Division of Virus-Associated Carcinogenesis”, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- IB-Cancer Research Foundation, Science Park 2, 66123 Saarbrücken, Germany
| | - Gael Palais
- German Cancer Research Center, “Division of Virus-Associated Carcinogenesis”, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Maria Qatato
- German Cancer Research Center, “Division of Virus-Associated Carcinogenesis”, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Maria Polycarpou-Schwarz
- German Cancer Research Center, “Division of Virus-Associated Carcinogenesis”, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Martin Schneider
- German Cancer Research Center, Mass Spectrometry based Protein Analysis Unit, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Christina Kalk
- German Cancer Research Center, “Division of Virus-Associated Carcinogenesis”, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Wibke Rüdiger
- German Cancer Research Center, “Division of Virus-Associated Carcinogenesis”, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Alina Ertl
- German Cancer Research Center, “Division of Virus-Associated Carcinogenesis”, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Natasha Anstee
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- German Cancer Research Center, “Division of Experimental Hematology”, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Ruzhica Bogeska
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- German Cancer Research Center, “Division of Experimental Hematology”, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Dominic Helm
- German Cancer Research Center, Mass Spectrometry based Protein Analysis Unit, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Michael D. Milsom
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- German Cancer Research Center, “Division of Experimental Hematology”, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Bruno Galy
- German Cancer Research Center, “Division of Virus-Associated Carcinogenesis”, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| |
Collapse
|
26
|
Wiencke JK, Molinaro AM, Warrier G, Rice T, Clarke J, Taylor JW, Wrensch M, Hansen H, McCoy L, Tang E, Tamaki SJ, Tamaki CM, Nissen E, Bracci P, Salas LA, Koestler DC, Christensen BC, Zhang Z, Kelsey KT. DNA methylation as a pharmacodynamic marker of glucocorticoid response and glioma survival. Nat Commun 2022; 13:5505. [PMID: 36127421 PMCID: PMC9486797 DOI: 10.1038/s41467-022-33215-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 09/08/2022] [Indexed: 12/15/2022] Open
Abstract
Assessing individual responses to glucocorticoid drug therapies that compromise immune status and affect survival outcomes in neuro-oncology is a great challenge. Here we introduce a blood-based neutrophil dexamethasone methylation index (NDMI) that provides a measure of the epigenetic response of subjects to dexamethasone. This marker outperforms conventional approaches based on leukocyte composition as a marker of glucocorticoid response. The NDMI is associated with low CD4 T cells and the accumulation of monocytic myeloid-derived suppressor cells and also serves as prognostic factor in glioma survival. In a non-glioma population, the NDMI increases with a history of prednisone use. Therefore, it may also be informative in other conditions where glucocorticoids are employed. We conclude that DNA methylation remodeling within the peripheral immune compartment is a rich source of clinically relevant markers of glucocorticoid response.
Collapse
Affiliation(s)
- J K Wiencke
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA.
| | - Annette M Molinaro
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Gayathri Warrier
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Terri Rice
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Jennifer Clarke
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Jennie W Taylor
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Margaret Wrensch
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Helen Hansen
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Lucie McCoy
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Emily Tang
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Stan J Tamaki
- Parnassus Flow Cytometry CoLab, University of California San Francisco, San Francisco, CA, USA
| | - Courtney M Tamaki
- Parnassus Flow Cytometry CoLab, University of California San Francisco, San Francisco, CA, USA
| | - Emily Nissen
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS, USA
| | - Paige Bracci
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Lucas A Salas
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | - Devin C Koestler
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS, USA
| | - Brock C Christensen
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
- Department of Molecular and Systems Biology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
- Department of Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | - Ze Zhang
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | - Karl T Kelsey
- Department of Epidemiology, Brown University, Providence, RI, USA
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| |
Collapse
|
27
|
Ortmayr K, Zampieri M. Sorting-free metabolic profiling uncovers the vulnerability of fatty acid β-oxidation in in vitro quiescence models. Mol Syst Biol 2022; 18:e10716. [PMID: 36094015 PMCID: PMC9465820 DOI: 10.15252/msb.202110716] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 08/18/2022] [Accepted: 08/22/2022] [Indexed: 11/09/2022] Open
Abstract
Quiescent cancer cells are rare nondiving cells with the unique ability to evade chemotherapies and resume cell division after treatment. Despite the associated risk of cancer recurrence, how cells can reversibly switch between rapid proliferation and quiescence remains a long-standing open question. By developing a unique methodology for the cell sorting-free separation of metabolic profiles in cell subpopulations in vitro, we unraveled metabolic characteristics of quiescent cells that are largely invariant to basal differences in cell types and quiescence-inducing stimuli. Consistent with our metabolome-based analysis, we show that impairing mitochondrial fatty acid β-oxidation (FAO) can induce apoptosis in quiescence-induced cells and hamper their return to proliferation. Our findings suggest that in addition to mediating energy and redox balance, FAO can play a role in preventing the buildup of toxic intermediates during transitioning to quiescence. Uncovering metabolic strategies to enter, maintain, and exit quiescence can reveal fundamental principles in cell plasticity and new potential therapeutic targets beyond cancer.
Collapse
Affiliation(s)
- Karin Ortmayr
- Institute of Molecular Systems Biology, ETHZürichSwitzerland
- Division of Pharmacognosy, Department of Pharmaceutical Sciences, Faculty of Life SciencesUniversity of ViennaViennaAustria
| | - Mattia Zampieri
- Institute of Molecular Systems Biology, ETHZürichSwitzerland
| |
Collapse
|
28
|
Overbeeke C, Tak T, Koenderman L. The journey of neutropoiesis: how complex landscapes in bone marrow guide continuous neutrophil lineage determination. Blood 2022; 139:2285-2293. [PMID: 34986245 PMCID: PMC11022826 DOI: 10.1182/blood.2021012835] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 12/31/2021] [Indexed: 12/16/2022] Open
Abstract
Neutrophils are the most abundant white blood cell, and they differentiate in homeostasis in the bone marrow from hematopoietic stem cells (HSCs) via multiple intermediate progenitor cells into mature cells that enter the circulation. Recent findings support a continuous model of differentiation in the bone marrow of heterogeneous HSCs and progenitor populations. Cell fate decisions at the levels of proliferation and differentiation are enforced through expression of lineage-determining transcription factors and their interactions, which are influenced by intrinsic (intracellular) and extrinsic (extracellular) mechanisms. Neutrophil homeostasis is subjected to positive-feedback loops, stemming from the gut microbiome, as well as negative-feedback loops resulting from the clearance of apoptotic neutrophils by mature macrophages. Finally, the cellular kinetics regarding the replenishing of the mature neutrophil pool is discussed in light of recent contradictory data.
Collapse
Affiliation(s)
- Celine Overbeeke
- Department of Respiratory Medicine and Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Tamar Tak
- Department of Parasitology, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Leo Koenderman
- Department of Respiratory Medicine and Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
29
|
Formation of neutrophil extracellular traps requires actin cytoskeleton rearrangements. Blood 2022; 139:3166-3180. [PMID: 35030250 DOI: 10.1182/blood.2021013565] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 01/09/2022] [Indexed: 11/20/2022] Open
Abstract
Neutrophils are important effector cells in the host defense against invading micro-organisms. One of the mechanisms they employ to eliminate pathogens is the release of neutrophil extracellular traps (NETs). Although NET release and subsequent cell death known as NETosis have been intensively studied, the cellular components and factors determining or facilitating the formation of NETs remain incompletely understood. Using various actin polymerization and myosin II modulators on neutrophils from healthy individuals, we show that intact F-actin dynamics and myosin II function are essential for NET formation when induced by different stimuli, i.e. phorbol 12-myristate 13-acetate, monosodium urate crystals and Candida albicans. The role of actin polymerization in NET formation could not be explained by the lack of reactive oxygen species production or granule release, which were normal or enhanced under the given conditions. Neutrophils from patients with very rare inherited actin polymerization defects by either ARPC1B- or MKL1-deficiency also failed to show NETosis. We found that upon inhibition of actin dynamics there is a lack of translocation of NE to the nucleus, which may well explain the impaired NET formation. Collectively, our data illustrate the essential requirement of an intact and active actin polymerization process, as well as active myosin II to enable the release of nuclear DNA by neutrophils during NET formation.
Collapse
|
30
|
S100A8/A9 Is a Marker for the Release of Neutrophil Extracellular Traps and Induces Neutrophil Activation. Cells 2022; 11:cells11020236. [PMID: 35053354 PMCID: PMC8773660 DOI: 10.3390/cells11020236] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/22/2021] [Accepted: 01/06/2022] [Indexed: 12/12/2022] Open
Abstract
Neutrophils are the most abundant innate immune cells in the circulation and they are the first cells recruited to sites of infection or inflammation. Almost half of the intracellular protein content in neutrophils consists of S100A8 and S100A9, though there has been controversy about their actual localization. Once released extracellularly, these proteins are thought to act as damage-associated molecular patterns (DAMPs), though their mechanism of action is not well understood. These S100 proteins mainly form heterodimers (S100A8/A9, also known as calprotectin) and this heterocomplex is recognized as a useful biomarker for several inflammatory diseases. We observed that S100A8/A9 is highly present in the cytoplasmic fraction of neutrophils and is not part of the granule content. Furthermore, we found that S100A8/A9 was not released in parallel with granular content but upon the formation of neutrophil extracellular traps (NETs). Accordingly, neutrophils of patients with chronic granulomatous disease, who are deficient in phorbol 12-myristate 13-acetate (PMA)-induced NETosis, did not release S100A8/A9 upon PMA stimulation. Moreover, we purified S100A8/A9 from the cytoplasmic fraction of neutrophils and found that S100A8/A9 could induce neutrophil activation, including adhesion and CD11b upregulation, indicating that this DAMP might amplify neutrophil activation.
Collapse
|
31
|
Ferragut Cardoso AP, Banerjee M, Al-Eryani L, Sayed M, Wilkey DW, Merchant ML, Park JW, States JC. Temporal Modulation of Differential Alternative Splicing in HaCaT Human Keratinocyte Cell Line Chronically Exposed to Arsenic for up to 28 Wk. ENVIRONMENTAL HEALTH PERSPECTIVES 2022; 130:17011. [PMID: 35072517 PMCID: PMC8785870 DOI: 10.1289/ehp9676] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
BACKGROUND Chronic arsenic exposure via drinking water is associated with an increased risk of developing cancer and noncancer chronic diseases. Pre-mRNAs are often subject to alternative splicing, generating mRNA isoforms encoding functionally distinct protein isoforms. The resulting imbalance in isoform species can result in pathogenic changes in critical signaling pathways. Alternative splicing as a mechanism of arsenic-induced toxicity and carcinogenicity is understudied. OBJECTIVE This study aimed to accurately profile differential alternative splicing events in human keratinocytes induced by chronic arsenic exposure that might play a role in carcinogenesis. METHODS Independent quadruplicate cultures of immortalized human keratinocytes (HaCaT) were maintained continuously for 28 wk with 0 or 100 nM sodium arsenite. RNA-sequencing (RNA-Seq) was performed with poly(A) RNA isolated from cells harvested at 7, 19, and 28 wk with subsequent replicate multivariate analysis of transcript splicing (rMATS) analysis to detect and quantify differential alternative splicing events. Reverse transcriptase-polymerase chain reaction (RT-PCR) for selected alternative splicing events was performed to validate RNA-Seq predictions. Functional enrichment was performed by gene ontology (GO) analysis of the differential alternative splicing event data set at each time point. RESULTS At least 600 differential alternative splicing events were detected at each time point tested, comprising all the five main types of alternative splicing and occurring in both open reading frames (ORFs) and untranslated regions (UTRs). Based on functional relevance ELK4, SHC1, and XRRA1 were selected for validation of predicted alternative splicing events at 7 wk by RT-PCR. Densitometric analysis of RT-PCR data corroborated the rMATS predicted alternative splicing for all three events. Protein expression validation of the selected alternative splicing events was challenging given that very few isoform-specific antibodies are available. GO analysis demonstrated that the enriched terms in differential alternatively spliced mRNAs changed dynamically with the time of exposure. Notably, RNA metabolism and splicing regulation pathways were enriched at the 7-wk time point, when the greatest number of differentially alternatively spliced mRNAs are detected. Our preliminary proteomic analysis demonstrated that the expression of the canonical isoforms of the splice regulators DDX42, RMB25, and SRRM2 were induced upon chronic arsenic exposure, corroborating the splicing predictions. DISCUSSION These results using cultures of HaCaT cells suggest that arsenic exposure disrupted an alternative splice factor network and induced time-dependent genome-wide differential alternative splicing that likely contributed to the changing proteomic landscape in arsenic-induced carcinogenesis. However, significant challenges remain in corroborating alternative splicing data at the proteomic level. https://doi.org/10.1289/EHP9676.
Collapse
Affiliation(s)
- Ana P. Ferragut Cardoso
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA
| | - Mayukh Banerjee
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA
| | - Laila Al-Eryani
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA
| | - Mohammed Sayed
- Computer Science and Engineering, University of Louisville, Louisville, Kentucky, USA
| | - Daniel W. Wilkey
- Division of Nephrology & Hypertension, Department of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Michael L. Merchant
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA
- Division of Nephrology & Hypertension, Department of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Juw W. Park
- Computer Science and Engineering, University of Louisville, Louisville, Kentucky, USA
- KY INBRE Bioinformatics Core, University of Louisville, Louisville, Kentucky, USA
| | - J. Christopher States
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
32
|
Ganopoulou M, Michailidis M, Angelis L, Ganopoulos I, Molassiotis A, Xanthopoulou A, Moysiadis T. Could Causal Discovery in Proteogenomics Assist in Understanding Gene-Protein Relations? A Perennial Fruit Tree Case Study Using Sweet Cherry as a Model. Cells 2021; 11:cells11010092. [PMID: 35011654 PMCID: PMC8750600 DOI: 10.3390/cells11010092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 12/12/2022] Open
Abstract
Genome-wide transcriptome analysis is a method that produces important data on plant biology at a systemic level. The lack of understanding of the relationships between proteins and genes in plants necessitates a further thorough analysis at the proteogenomic level. Recently, our group generated a quantitative proteogenomic atlas of 15 sweet cherry (Prunus avium L.) cv. ‘Tragana Edessis’ tissues represented by 29,247 genes and 7584 proteins. The aim of the current study was to perform a targeted analysis at the gene/protein level to assess the structure of their relation, and the biological implications. Weighted correlation network analysis and causal modeling were employed to, respectively, cluster the gene/protein pairs, and reveal their cause–effect relations, aiming to assess the associated biological functions. To the best of our knowledge, this is the first time that causal modeling has been employed within the proteogenomics concept in plants. The analysis revealed the complex nature of causal relations among genes/proteins that are important for traits of interest in perennial fruit trees, particularly regarding the fruit softening and ripening process in sweet cherry. Causal discovery could be used to highlight persistent relations at the gene/protein level, stimulating biological interpretation and facilitating further study of the proteogenomic atlas in plants.
Collapse
Affiliation(s)
- Maria Ganopoulou
- School of Informatics, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
- Correspondence: (M.G.); (T.M.)
| | - Michail Michailidis
- Laboratory of Pomology, Department of Horticulture, Aristotle University of Thessaloniki, Thermi, 57001 Thessaloniki, Greece; (M.M.); (A.M.); (A.X.)
| | - Lefteris Angelis
- School of Informatics, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Ioannis Ganopoulos
- Institute of Plant Breeding and Genetic Resources, ELGO-DIMITRA, Thermi, 57001 Thessaloniki, Greece;
| | - Athanassios Molassiotis
- Laboratory of Pomology, Department of Horticulture, Aristotle University of Thessaloniki, Thermi, 57001 Thessaloniki, Greece; (M.M.); (A.M.); (A.X.)
| | - Aliki Xanthopoulou
- Laboratory of Pomology, Department of Horticulture, Aristotle University of Thessaloniki, Thermi, 57001 Thessaloniki, Greece; (M.M.); (A.M.); (A.X.)
- Institute of Plant Breeding and Genetic Resources, ELGO-DIMITRA, Thermi, 57001 Thessaloniki, Greece;
| | - Theodoros Moysiadis
- Institute of Plant Breeding and Genetic Resources, ELGO-DIMITRA, Thermi, 57001 Thessaloniki, Greece;
- Department of Computer Science, School of Sciences and Engineering, University of Nicosia, Nicosia 2417, Cyprus
- Correspondence: (M.G.); (T.M.)
| |
Collapse
|
33
|
Fisher J, Kahn F, Wiebe E, Gustafsson P, Kander T, Mellhammar L, Bentzer P, Linder A. The Dynamics of Circulating Heparin-Binding Protein: Implications for Its Use as a Biomarker. J Innate Immun 2021; 14:447-460. [PMID: 34965528 PMCID: PMC9485916 DOI: 10.1159/000521064] [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: 09/17/2021] [Accepted: 11/12/2021] [Indexed: 11/19/2022] Open
Abstract
Heparin-binding protein (HBP) is a promising biomarker for the development and severity of sepsis. To guide its use, it is important to understand the factors that could lead to false-positive or negative results, such as inappropriate release and inadequate clearance of HBP. HBP is presumably released only by neutrophils, and the organs responsible for its elimination are unknown. In this study, we aimed to determine whether non-neutrophil cells can be a source of circulating HBP and which organs are responsible for its removal. We found that in two cohorts of neutropenic patients, 12% and 19% of patients in each cohort, respectively, had detectable plasma HBP levels. In vitro, three leukemia-derived monocytic cell lines and healthy CD14+ monocytes constitutively released detectable levels of HBP. When HBP was injected intravenously in rats, we found that plasma levels of HBP decreased rapidly, with a distribution half-life below 10 min and an elimination half-life of 1-2 h. We measured HBP levels in the liver, spleen, kidneys, lungs, and urine using both ELISA and immunofluorescence quantitation, and found that the majority of HBP was present in the liver, and a small amount was present in the spleen. Immunofluorescence imaging indicated that HBP is associated mainly with hepatocytes in the liver and monocytes/macrophages in the spleen. The impact of hematologic malignancies and liver diseases on plasma HBP levels should be explored further in clinical studies.
Collapse
Affiliation(s)
- Jane Fisher
- Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Fredrik Kahn
- Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Elena Wiebe
- Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden.,Infection Biochemistry & Institute for Biochemistry, University of Veterinary Medicine Hanover, Hanover, Germany
| | - Pontus Gustafsson
- Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Thomas Kander
- Department of Intensive and Perioperative Care, Skåne University Hospital, Lund, Sweden.,Division of Anesthesiology and Intensive Care, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Lisa Mellhammar
- Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Peter Bentzer
- Division of Anesthesiology and Intensive Care, Department of Clinical Sciences Lund, Lund University, Lund, Sweden.,Department of Anesthesia and Intensive Care, Helsingborg Hospital, Helsingborg, Sweden
| | - Adam Linder
- Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| |
Collapse
|
34
|
Fox E, Jones R, Samanta R, Summers C. Characterising the transcriptome of hypersegmented human neutrophils. Wellcome Open Res 2021. [DOI: 10.12688/wellcomeopenres.17440.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background: Mature human neutrophils are characterised by their multilobed nuclear morphology. Neutrophil hypersegmentation, a pathologic nuclear phenotype, has been described in the alveolar compartment of patients with acute respiratory distress syndrome and in several other contexts. This study aimed to characterise the transcriptional changes associated with neutrophil hypersegmentation. Methods: A model of hypersegmentation was established by exposing healthy peripheral blood neutrophils to the angiotensin converting enzyme inhibitor (ACEi) captopril. Laser capture microdissection (LCM) was then adapted to isolate a population of hypersegmented neutrophils. Transcriptomic analysis of microdissected hypersegmented neutrophils was undertaken using ribonucleic acid (RNA) sequencing. Differential gene expression (DEG) and enrichment pathway analysis were conducted to investigate the mechanisms underlying hypersegmentation. Results: RNA-Seq analysis revealed the transcriptomic signature of hypersegmented neutrophils, with five genes differentially expressed. VCAN, PADI4 and DUSP4 were downregulated, while LTF and PSMC4 were upregulated. Modulated pathways included histone modification, protein-DNA complex assembly and antimicrobial humoral response. The role of PADI4 was further validated using the small molecule inhibitor, Cl-amidine. Conclusions: Hypersegmented neutrophils display a marked transcriptomic signature, characterised by the differential expression of five genes. This study provides insights into the mechanisms underlying neutrophil hypersegmentation and describes a novel method to isolate and sequence neutrophils based on their morphologic subtype.
Collapse
|
35
|
Taniguchi-Ponciano K, Vadillo E, Mayani H, Gonzalez-Bonilla CR, Torres J, Majluf A, Flores-Padilla G, Wacher-Rodarte N, Galan JC, Ferat-Osorio E, Blanco-Favela F, Lopez-Macias C, Ferreira-Hermosillo A, Ramirez-Renteria C, Peña-Martínez E, Silva-Román G, Vela-Patiño S, Mata-Lozano C, Carvente-Garcia R, Basurto-Acevedo L, Saucedo R, Piña-Sanchez P, Chavez-Gonzalez A, Marrero-Rodríguez D, Mercado M. Increased expression of hypoxia-induced factor 1α mRNA and its related genes in myeloid blood cells from critically ill COVID-19 patients. Ann Med 2021; 53:197-207. [PMID: 33345622 PMCID: PMC7784832 DOI: 10.1080/07853890.2020.1858234] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 11/25/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND COVID-19 counts 46 million people infected and killed more than 1.2 million. Hypoxaemia is one of the main clinical manifestations, especially in severe cases. HIF1α is a master transcription factor involved in the cellular response to oxygen levels. The immunopathogenesis of this severe form of COVID-19 is poorly understood. METHODS We performed scRNAseq from leukocytes from five critically ill COVID-19 patients and characterized the expression of hypoxia-inducible factor1α and its transcriptionally regulated genes. Also performed metanalysis from the publicly available RNAseq data from COVID-19 bronchoalveolar cells. RESULTS Critically-ill COVID-19 patients show a shift towards an immature myeloid profile in peripheral blood cells, including band neutrophils, immature monocytes, metamyelocytes, monocyte-macrophages, monocytoid precursors, and promyelocytes-myelocytes, together with mature monocytes and segmented neutrophils. May be the result of a physiological response known as emergency myelopoiesis. These cellular subsets and bronchoalveolar cells express HIF1α and their transcriptional targets related to inflammation (CXCL8, CXCR1, CXCR2, and CXCR4); virus sensing, (TLR2 and TLR4); and metabolism (SLC2A3, PFKFB3, PGK1, GAPDH and SOD2). CONCLUSIONS The up-regulation and participation of HIF1α in events such as inflammation, immunometabolism, and TLR make it a potential molecular marker for COVID-19 severity and, interestingly, could represent a potential target for molecular therapy. Key messages Critically ill COVID-19 patients show emergency myelopoiesis. HIF1α and its transcriptionally regulated genes are expressed in immature myeloid cells which could serve as molecular targets. HIF1α and its transcriptionally regulated genes is also expressed in lung cells from critically ill COVID-19 patients which may partially explain the hypoxia related events.
Collapse
Affiliation(s)
- Keiko Taniguchi-Ponciano
- Unidad de Investigación Médica en Enfermedades Endocrinas, UMAE Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico city, Mexico
| | - Eduardo Vadillo
- Unidad de Investigación Médica en Enfermedades Oncológicas, UMAE Hospital de Oncología, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico city, Mexico
| | - Héctor Mayani
- Unidad de Investigación Médica en Enfermedades Oncológicas, UMAE Hospital de Oncología, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico city, Mexico
| | | | - Javier Torres
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, UMAE Hospital de Pediatría, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico city, Mexico
| | - Abraham Majluf
- Unidad de Investigación Médica en trombosis, hemostasia y aterogénesis, Instituto Mexicano del Seguro Social, Mexico city, Mexico
| | - Guillermo Flores-Padilla
- Servicio de Medicina Interna, UMAE Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico city, Mexico
| | - Niels Wacher-Rodarte
- Unidad de Investigación Médica en Epidemiología Clínica, UMAE Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico city, Mexico
| | - Juan Carlos Galan
- Servicio de Medicina Interna, UMAE Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico city, Mexico
| | - Eduardo Ferat-Osorio
- División de Investigación en Salud, UMAE Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico city, Mexico
| | - Francisco Blanco-Favela
- Unidad de Investigación Médica en Inmunología, UMAE Hospital de Pediatría, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico city, Mexico
| | - Constantino Lopez-Macias
- Unidad de Investigación Médica en Inmunoquímica, UMAE Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico city, Mexico
| | - Aldo Ferreira-Hermosillo
- Unidad de Investigación Médica en Enfermedades Endocrinas, UMAE Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico city, Mexico
| | - Claudia Ramirez-Renteria
- Unidad de Investigación Médica en Enfermedades Endocrinas, UMAE Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico city, Mexico
| | - Eduardo Peña-Martínez
- Unidad de Investigación Médica en Enfermedades Endocrinas, UMAE Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico city, Mexico
| | - Gloria Silva-Román
- Unidad de Investigación Médica en Enfermedades Endocrinas, UMAE Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico city, Mexico
| | - Sandra Vela-Patiño
- Unidad de Investigación Médica en Enfermedades Endocrinas, UMAE Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico city, Mexico
| | - Carlos Mata-Lozano
- Unidad de Investigación Médica en Enfermedades Endocrinas, UMAE Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico city, Mexico
- Analitek S.A. de C.V., CDMX, México
| | - Roberto Carvente-Garcia
- Unidad de Investigación Médica en Enfermedades Endocrinas, UMAE Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico city, Mexico
- Analitek S.A. de C.V., CDMX, México
| | - Lourdes Basurto-Acevedo
- Unidad de Investigación Médica en Enfermedades Endocrinas, UMAE Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico city, Mexico
| | - Renata Saucedo
- Unidad de Investigación Médica en Enfermedades Endocrinas, UMAE Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico city, Mexico
| | - Patricia Piña-Sanchez
- Unidad de Investigación Médica en Enfermedades Oncológicas, UMAE Hospital de Oncología, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico city, Mexico
| | - Antonieta Chavez-Gonzalez
- Unidad de Investigación Médica en Enfermedades Oncológicas, UMAE Hospital de Oncología, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico city, Mexico
| | - Daniel Marrero-Rodríguez
- Unidad de Investigación Médica en Enfermedades Endocrinas, UMAE Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico city, Mexico
| | - Moisés Mercado
- Unidad de Investigación Médica en Enfermedades Endocrinas, UMAE Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico city, Mexico
| |
Collapse
|
36
|
Harnik Y, Buchauer L, Ben-Moshe S, Averbukh I, Levin Y, Savidor A, Eilam R, Moor AE, Itzkovitz S. Spatial discordances between mRNAs and proteins in the intestinal epithelium. Nat Metab 2021; 3:1680-1693. [PMID: 34931081 DOI: 10.1038/s42255-021-00504-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 11/05/2021] [Indexed: 12/14/2022]
Abstract
The use of transcriptomes as reliable proxies for cellular proteomes is controversial. In the small intestine, enterocytes operate for 4 days as they migrate along villi, which are highly graded microenvironments. Spatial transcriptomics have demonstrated profound zonation in enterocyte gene expression, but how this variability translates to protein content is unclear. Here we show that enterocyte proteins and messenger RNAs along the villus axis are zonated, yet often spatially discordant. Using spatial sorting with zonated surface markers, together with a Bayesian approach to infer protein translation and degradation rates from the combined spatial profiles, we find that, while many genes exhibit proteins zonated toward the villus tip, mRNA is zonated toward the villus bottom. Finally, we demonstrate that space-independent protein synthesis delays can explain many of the mRNA-protein discordances. Our work provides a proteomic spatial blueprint of the intestinal epithelium, highlighting the importance of protein measurements for inferring cell states in tissues that operate outside of steady state.
Collapse
Affiliation(s)
- Yotam Harnik
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Lisa Buchauer
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Shani Ben-Moshe
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Inna Averbukh
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Yishai Levin
- The De Botton Institute for Protein Profiling, The Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Alon Savidor
- The De Botton Institute for Protein Profiling, The Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Raya Eilam
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Andreas E Moor
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Shalev Itzkovitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
37
|
Mocciaro E, Runfola V, Ghezzi P, Pannese M, Gabellini D. DUX4 Role in Normal Physiology and in FSHD Muscular Dystrophy. Cells 2021; 10:3322. [PMID: 34943834 PMCID: PMC8699294 DOI: 10.3390/cells10123322] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/10/2021] [Accepted: 11/23/2021] [Indexed: 12/12/2022] Open
Abstract
In the last decade, the sequence-specific transcription factor double homeobox 4 (DUX4) has gone from being an obscure entity to being a key factor in important physiological and pathological processes. We now know that expression of DUX4 is highly regulated and restricted to the early steps of embryonic development, where DUX4 is involved in transcriptional activation of the zygotic genome. While DUX4 is epigenetically silenced in most somatic tissues of healthy humans, its aberrant reactivation is associated with several diseases, including cancer, viral infection and facioscapulohumeral muscular dystrophy (FSHD). DUX4 is also translocated, giving rise to chimeric oncogenic proteins at the basis of sarcoma and leukemia forms. Hence, understanding how DUX4 is regulated and performs its activity could provide relevant information, not only to further our knowledge of human embryonic development regulation, but also to develop therapeutic approaches for the diseases associated with DUX4. Here, we summarize current knowledge on the cellular and molecular processes regulated by DUX4 with a special emphasis on FSHD muscular dystrophy.
Collapse
Affiliation(s)
| | | | | | | | - Davide Gabellini
- Gene Expression and Muscular Dystrophy Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, 20132 Milano, Italy; (E.M.); (V.R.); (P.G.); (M.P.)
| |
Collapse
|
38
|
Sollberger G. Approaching Neutrophil Pyroptosis. J Mol Biol 2021; 434:167335. [PMID: 34757055 DOI: 10.1016/j.jmb.2021.167335] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 01/21/2023]
Abstract
All cells must die at some point, and the dogma is that they do it either silently via apoptosis or via pro-inflammatory, lytic forms of death. Amongst these lytic cell death pathways, pyroptosis is one of the best characterized. Pyroptosis depends on inflammatory caspases which activate members of the gasdermin family of proteins, and it is associated with the release of the pro-inflammatory cytokines interleukin (IL)-1β and IL-18. Pyroptosis is an essential component of innate immunity, it initiates and amplifies inflammation and it removes the replication niche for intracellular pathogens. Most of the literature on pyroptosis focuses on monocytes and macrophages. However, the most abundant phagocytes in humans are neutrophils. This review addresses whether neutrophils undergo pyroptosis and the underlying mechanisms. Furthermore, I discuss how and why neutrophils might be able to resist pyroptosis.
Collapse
Affiliation(s)
- Gabriel Sollberger
- University of Dundee, School of Life Sciences, Division of Cell Signalling and Immunology, Dow Street, DD1 5EH Dundee, UK; Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
39
|
Yu S, Drton M, Promislow DEL, Shojaie A. CorDiffViz: an R package for visualizing multi-omics differential correlation networks. BMC Bioinformatics 2021; 22:486. [PMID: 34627139 PMCID: PMC8501646 DOI: 10.1186/s12859-021-04383-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 09/20/2021] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Differential correlation networks are increasingly used to delineate changes in interactions among biomolecules. They characterize differences between omics networks under two different conditions, and can be used to delineate mechanisms of disease initiation and progression. RESULTS We present a new R package, CorDiffViz, that facilitates the estimation and visualization of differential correlation networks using multiple correlation measures and inference methods. The software is implemented in R, HTML and Javascript, and is available at https://github.com/sqyu/CorDiffViz . Visualization has been tested for the Chrome and Firefox web browsers. A demo is available at https://diffcornet.github.io/CorDiffViz/demo.html . CONCLUSIONS Our software offers considerable flexibility by allowing the user to interact with the visualization and choose from different estimation methods and visualizations. It also allows the user to easily toggle between correlation networks for samples under one condition and differential correlations between samples under two conditions. Moreover, the software facilitates integrative analysis of cross-correlation networks between two omics data sets.
Collapse
Affiliation(s)
- Shiqing Yu
- Department of Statistics, University of Washington, NE Stevens Way, Seattle, WA, 98195, USA.
| | - Mathias Drton
- Department of Mathematics, Technical University of Munich, Boltzmannstraße, 85748, Garching bei München, Germany
| | - Daniel E L Promislow
- Departments of Pathology and Biology, University of Washington, NE Pacific St, Seattle, WA, 98195, USA
| | - Ali Shojaie
- Department of Biostatistics, University of Washington, NE Pacific St, Seattle, WA, 98195, USA
| |
Collapse
|
40
|
Kaiser R, Leunig A, Pekayvaz K, Popp O, Joppich M, Polewka V, Escaig R, Anjum A, Hoffknecht ML, Gold C, Brambs S, Engel A, Stockhausen S, Knottenberg V, Titova A, Haji M, Scherer C, Muenchhoff M, Hellmuth JC, Saar K, Schubert B, Hilgendorff A, Schulz C, Kääb S, Zimmer R, Hübner N, Massberg S, Mertins P, Nicolai L, Stark K. Self-sustaining IL-8 loops drive a prothrombotic neutrophil phenotype in severe COVID-19. JCI Insight 2021; 6:e150862. [PMID: 34403366 PMCID: PMC8492337 DOI: 10.1172/jci.insight.150862] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 08/11/2021] [Indexed: 12/15/2022] Open
Abstract
Neutrophils provide a critical line of defense in immune responses to various pathogens, inflicting self-damage upon transition to a hyperactivated, procoagulant state. Recent work has highlighted proinflammatory neutrophil phenotypes contributing to lung injury and acute respiratory distress syndrome (ARDS) in patients with coronavirus disease 2019 (COVID-19). Here, we use state-of-the art mass spectrometry-based proteomics and transcriptomic and correlative analyses as well as functional in vitro and in vivo studies to dissect how neutrophils contribute to the progression to severe COVID-19. We identify a reinforcing loop of both systemic and neutrophil intrinsic IL-8 (CXCL8/IL-8) dysregulation, which initiates and perpetuates neutrophil-driven immunopathology. This positive feedback loop of systemic and neutrophil autocrine IL-8 production leads to an activated, prothrombotic neutrophil phenotype characterized by degranulation and neutrophil extracellular trap (NET) formation. In severe COVID-19, neutrophils directly initiate the coagulation and complement cascade, highlighting a link to the immunothrombotic state observed in these patients. Targeting the IL-8-CXCR-1/-2 axis interferes with this vicious cycle and attenuates neutrophil activation, degranulation, NETosis, and IL-8 release. Finally, we show that blocking IL-8-like signaling reduces severe acute respiratory distress syndrome of coronavirus 2 (SARS-CoV-2) spike protein-induced, human ACE2-dependent pulmonary microthrombosis in mice. In summary, our data provide comprehensive insights into the activation mechanisms of neutrophils in COVID-19 and uncover a self-sustaining neutrophil-IL-8 axis as a promising therapeutic target in severe SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Rainer Kaiser
- Department of Medicine I, University Hospital, Ludwig-Maximilians University Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- COVID-19 Registry of the LMU Munich (CORKUM), University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Alexander Leunig
- Department of Medicine I, University Hospital, Ludwig-Maximilians University Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Kami Pekayvaz
- Department of Medicine I, University Hospital, Ludwig-Maximilians University Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- COVID-19 Registry of the LMU Munich (CORKUM), University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Oliver Popp
- Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
- DZHK, partner site Berlin, Berlin, Germany
| | - Markus Joppich
- Department of Informatics, Ludwig-Maximilians University Munich, Munich, Germany
| | - Vivien Polewka
- Department of Medicine I, University Hospital, Ludwig-Maximilians University Munich, Germany
| | - Raphael Escaig
- Department of Medicine I, University Hospital, Ludwig-Maximilians University Munich, Germany
| | - Afra Anjum
- Department of Medicine I, University Hospital, Ludwig-Maximilians University Munich, Germany
| | - Marie-Louise Hoffknecht
- Department of Medicine I, University Hospital, Ludwig-Maximilians University Munich, Germany
| | - Christoph Gold
- Department of Medicine I, University Hospital, Ludwig-Maximilians University Munich, Germany
| | - Sophia Brambs
- Department of Medicine I, University Hospital, Ludwig-Maximilians University Munich, Germany
| | - Anouk Engel
- Department of Medicine I, University Hospital, Ludwig-Maximilians University Munich, Germany
| | - Sven Stockhausen
- Department of Medicine I, University Hospital, Ludwig-Maximilians University Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Viktoria Knottenberg
- Department of Medicine I, University Hospital, Ludwig-Maximilians University Munich, Germany
| | - Anna Titova
- Department of Medicine I, University Hospital, Ludwig-Maximilians University Munich, Germany
| | - Mohamed Haji
- Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
- DZHK, partner site Berlin, Berlin, Germany
| | - Clemens Scherer
- Department of Medicine I, University Hospital, Ludwig-Maximilians University Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- COVID-19 Registry of the LMU Munich (CORKUM), University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Maximilian Muenchhoff
- COVID-19 Registry of the LMU Munich (CORKUM), University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
- Max von Pettenkofer Institute and GeneCenter, Virology, Faculty of Medicine, Ludwig-Maximilians University, Munich, Germany
- German Center for Infection Research, Partner Site Munich, Munich, Germany
| | - Johannes C. Hellmuth
- COVID-19 Registry of the LMU Munich (CORKUM), University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
- Medical Clinic and Polyclinic III, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Kathrin Saar
- Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
- DZHK, partner site Berlin, Berlin, Germany
| | - Benjamin Schubert
- Institute of Computational Biology, Helmholtz Zentrum München (German Research Center for Environmental Health), Neuherberg, Germany
- Department of Mathematics, Technical University of Munich, Garching, Germany
- The COMBAT C19IR study group is detailed in the Acknowledgments
| | - Anne Hilgendorff
- The COMBAT C19IR study group is detailed in the Acknowledgments
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Center Munich, Member of the German Center for Lung Research, Munich, Germany
- Center for Comprehensive Developmental Care at the interdisciplinary Social Pediatric Center, Haunersches Children’s Hospital, University Hospital Ludwig-Maximilian University, Munich, Germany
| | - Christian Schulz
- Department of Medicine I, University Hospital, Ludwig-Maximilians University Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Stefan Kääb
- Department of Medicine I, University Hospital, Ludwig-Maximilians University Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- COVID-19 Registry of the LMU Munich (CORKUM), University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Ralf Zimmer
- Department of Informatics, Ludwig-Maximilians University Munich, Munich, Germany
| | - Norbert Hübner
- Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
- DZHK, partner site Berlin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Germany
| | - Steffen Massberg
- Department of Medicine I, University Hospital, Ludwig-Maximilians University Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- COVID-19 Registry of the LMU Munich (CORKUM), University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Philipp Mertins
- Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
- DZHK, partner site Berlin, Berlin, Germany
| | - Leo Nicolai
- Department of Medicine I, University Hospital, Ludwig-Maximilians University Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- COVID-19 Registry of the LMU Munich (CORKUM), University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Konstantin Stark
- Department of Medicine I, University Hospital, Ludwig-Maximilians University Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- COVID-19 Registry of the LMU Munich (CORKUM), University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| |
Collapse
|
41
|
Current Understanding of the Neutrophil Transcriptome in Health and Disease. Cells 2021; 10:cells10092406. [PMID: 34572056 PMCID: PMC8469435 DOI: 10.3390/cells10092406] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 12/23/2022] Open
Abstract
Neutrophils are key cells of the innate immune system. It is now understood that this leukocyte population is diverse in both the basal composition and functional plasticity. Underlying this plasticity is a post-translational framework for rapidly achieving early activation states, but also a transcriptional capacity that is becoming increasingly recognized by immunologists. Growing interest in the contribution of neutrophils to health and disease has resulted in more efforts to describe their transcriptional activity. Whilst initial efforts focused predominantly on understanding the existing biology, investigations with advanced methods such as single cell RNA sequencing to understand interactions of the entire immune system are revealing higher flexibility in neutrophil transcription than previously thought possible and multiple transition states. It is now apparent that neutrophils utilise many forms of RNA in the regulation of their function. This review collates current knowledge on the nuclei structure and gene expression activity of human neutrophils across homeostasis and disease, before highlighting knowledge gaps that are research priority areas.
Collapse
|
42
|
Boschetti E, Zilberstein G, Righetti PG. Combinatorial peptides: A library that continuously probes low-abundance proteins. Electrophoresis 2021; 43:355-369. [PMID: 34498305 DOI: 10.1002/elps.202100131] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/31/2021] [Accepted: 08/24/2021] [Indexed: 12/12/2022]
Abstract
After a decade of experimental applications, it is the objective of this review to make a point on combinatorial peptide ligand libraries dedicated to low-abundance proteins from animals to plants and to microorganism proteomics. It is, thus, at the light of the recent technical developments and applications that we will examine the state of the art, its usage within the scientific community, and its openness to unexplored fields. The improvements of the methodology and its implementation in connection with analytical determinations of combinatorial peptide ligand library (CPLL)-treated samples are extensively reviewed and commented upon. Relevant examples covering few critical aspects describe the performance of the technology. Finally, a reflection on the technological future is attempted in particular by involving new concepts adapted to the limited availability of certain biological samples.
Collapse
Affiliation(s)
| | | | - Pier Giorgio Righetti
- Department of Chemistry Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan, Italy
| |
Collapse
|
43
|
Williams DW, Greenwell-Wild T, Brenchley L, Dutzan N, Overmiller A, Sawaya AP, Webb S, Martin D, Hajishengallis G, Divaris K, Morasso M, Haniffa M, Moutsopoulos NM. Human oral mucosa cell atlas reveals a stromal-neutrophil axis regulating tissue immunity. Cell 2021; 184:4090-4104.e15. [PMID: 34129837 PMCID: PMC8359928 DOI: 10.1016/j.cell.2021.05.013] [Citation(s) in RCA: 184] [Impact Index Per Article: 61.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/10/2021] [Accepted: 05/10/2021] [Indexed: 12/21/2022]
Abstract
The oral mucosa remains an understudied barrier tissue. This is a site of rich exposure to antigens and commensals, and a tissue susceptible to one of the most prevalent human inflammatory diseases, periodontitis. To aid in understanding tissue-specific pathophysiology, we compile a single-cell transcriptome atlas of human oral mucosa in healthy individuals and patients with periodontitis. We uncover the complex cellular landscape of oral mucosal tissues and identify epithelial and stromal cell populations with inflammatory signatures that promote antimicrobial defenses and neutrophil recruitment. Our findings link exaggerated stromal cell responsiveness with enhanced neutrophil and leukocyte infiltration in periodontitis. Our work provides a resource characterizing the role of tissue stroma in regulating mucosal tissue homeostasis and disease pathogenesis.
Collapse
Affiliation(s)
- Drake Winslow Williams
- Oral Immunity and Inflammation Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Teresa Greenwell-Wild
- Oral Immunity and Inflammation Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Laurie Brenchley
- Oral Immunity and Inflammation Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicolas Dutzan
- Oral Immunity and Inflammation Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA; Faculty of Dentistry, University of Chile, Santiago, Chile
| | - Andrew Overmiller
- Laboratory of Skin Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, MD 20892, USA
| | - Andrew Phillip Sawaya
- Laboratory of Skin Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, MD 20892, USA
| | - Simone Webb
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Daniel Martin
- Genomics and Computational Biology Core, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| | - George Hajishengallis
- University of Pennsylvania, Penn Dental Medicine, Department of Basic and Translational Sciences, Philadelphia, PA 19104, USA
| | - Kimon Divaris
- UNC Adams School of Dentistry and Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Maria Morasso
- Laboratory of Skin Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, MD 20892, USA
| | - Muzlifah Haniffa
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, UK; Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne NE2 4LP, UK
| | - Niki Maria Moutsopoulos
- Oral Immunity and Inflammation Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
44
|
Root L, Campo A, MacNiven L, Con P, Cnaani A, Kültz D. Nonlinear effects of environmental salinity on the gill transcriptome versus proteome of Oreochromis niloticus modulate epithelial cell turnover. Genomics 2021; 113:3235-3249. [PMID: 34298068 DOI: 10.1016/j.ygeno.2021.07.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/25/2021] [Accepted: 07/14/2021] [Indexed: 12/27/2022]
Abstract
A data-independent acquisition (DIA) assay library for targeted quantitation of thousands of Oreochromis niloticus gill proteins using a label- and gel-free workflow was generated and used to compare protein and mRNA abundances. This approach generated complimentary rather than redundant data for 1899 unique genes in gills of tilapia acclimated to freshwater and brackish water. Functional enrichment analyses identified mitochondrial energy metabolism, serine protease and immunity-related functions, and cytoskeleton/ extracellular matrix organization as major processes controlled by salinity in O. niloticus gills. Non-linearity in salinity-dependent transcriptome versus proteome regulation was revealed for specific functional groups of genes. The relationship was more linear for other molecular functions/ cellular processes, suggesting that the salinity-dependent regulation of O. niloticus gill function relies on post-transcriptional mechanisms for some functions/ processes more than others. This integrative systems biology approach can be adopted for other tissues and organisms to study cellular dynamics for many changing ecological contexts.
Collapse
Affiliation(s)
- Larken Root
- Department of Animal Sciences, University of California Davis, Meyer Hall, One Shields Avenue, Davis, CA 95616, USA
| | - Aurora Campo
- Department of Poultry and Aquaculture, Institute of Animal Sciences, Agricultural Research Organization, Volcani Center, P.O. Box 15159, Rishon LeZion 7528809, Israel
| | - Leah MacNiven
- Department of Animal Sciences, University of California Davis, Meyer Hall, One Shields Avenue, Davis, CA 95616, USA
| | - Pazit Con
- Department of Poultry and Aquaculture, Institute of Animal Sciences, Agricultural Research Organization, Volcani Center, P.O. Box 15159, Rishon LeZion 7528809, Israel
| | - Avner Cnaani
- Department of Poultry and Aquaculture, Institute of Animal Sciences, Agricultural Research Organization, Volcani Center, P.O. Box 15159, Rishon LeZion 7528809, Israel
| | - Dietmar Kültz
- Department of Animal Sciences, University of California Davis, Meyer Hall, One Shields Avenue, Davis, CA 95616, USA.
| |
Collapse
|
45
|
Neutrophil specific granule and NETosis defects in gray platelet syndrome. Blood Adv 2021; 5:549-564. [PMID: 33496751 DOI: 10.1182/bloodadvances.2020002442] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 12/06/2020] [Indexed: 12/15/2022] Open
Abstract
Gray platelet syndrome (GPS) is an autosomal recessive bleeding disorder characterized by a lack of α-granules in platelets and progressive myelofibrosis. Rare loss-of-function variants in neurobeachin-like 2 (NBEAL2), a member of the family of beige and Chédiak-Higashi (BEACH) genes, are causal of GPS. It is suggested that BEACH domain containing proteins are involved in fusion, fission, and trafficking of vesicles and granules. Studies in knockout mice suggest that NBEAL2 may control the formation and retention of granules in neutrophils. We found that neutrophils obtained from the peripheral blood from 13 patients with GPS have a normal distribution of azurophilic granules but show a deficiency of specific granules (SGs), as confirmed by immunoelectron microscopy and mass spectrometry proteomics analyses. CD34+ hematopoietic stem cells (HSCs) from patients with GPS differentiated into mature neutrophils also lacked NBEAL2 expression but showed similar SG protein expression as control cells. This is indicative of normal granulopoiesis in GPS and identifies NBEAL2 as a potentially important regulator of granule release. Patient neutrophil functions, including production of reactive oxygen species, chemotaxis, and killing of bacteria and fungi, were intact. NETosis was absent in circulating GPS neutrophils. Lack of NETosis is suggested to be independent of NBEAL2 expression but associated with SG defects instead, as indicated by comparison with HSC-derived neutrophils. Since patients with GPS do not excessively suffer from infections, the consequence of the reduced SG content and lack of NETosis for innate immunity remains to be explored.
Collapse
|
46
|
Comprehensive proteomic analysis of murine terminal erythroid differentiation. Blood Adv 2021; 4:1464-1477. [PMID: 32282884 DOI: 10.1182/bloodadvances.2020001652] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 03/04/2020] [Indexed: 12/12/2022] Open
Abstract
Murine-based cellular models have provided and continue to provide many useful insights into the fundamental mechanisms of erythropoiesis, as well as insights into the pathophysiology of inherited and acquired red cell disorders. Although detailed information on many aspects of these cell models is available, comprehensive proteomic data are lacking. This is a critical knowledge gap, as proteins are effectors of most biologic processes. To address this critical unmet need, proteomes of the murine cell lines Friend erythroleukemia (MEL), GATA1 erythroid (G1ER), and embryonic stem cell-derived erythroid progenitor (MEDEP) and proteomes of cultured murine marrow-derived erythroblasts at different stages of terminal erythroid differentiation were analyzed. The proteomes of MEDEP cells and primary murine erythroid cells were most similar, whereas those of MEL and G1ER cells were more distantly related. We demonstrated that the overall cellular content of histones does not decrease during terminal differentiation, despite strong chromatin condensation. Comparison of murine and human proteomes throughout terminal erythroid differentiation revealed that many noted transcriptomic changes were significantly dampened at the proteome level, especially at the end of the terminal differentiation process. Analysis of the early events associated with induction of terminal differentiation in MEDEP cells revealed divergent alterations in associated transcriptomes and proteomes. These proteomic data are powerful and valuable tools for the study of fundamental mechanisms of normal and disordered erythropoiesis and will be of broad interest to a wide range of investigators for making the appropriate choice of various cell lines to study inherited and acquired diseases of the erythrocyte.
Collapse
|
47
|
Abstract
INTRODUCTION The continuous technical improvement in sensitivity and specificity placed mass spectrometry as an alternative method for analyzing clinical samples. In parallel to the rapid development of discovery proteomics, targeted acquisition has been implemented as a complementary option for measuring a small set of proteins with high sensitivity and robustness in a large sample cohort. The combination of trapped ion mobility with a rapid time-of-flight (TOF) mass spectrometer improves the sensitivity even further and triggers the development of prm-PASEF. AREAS COVERED This article discusses the development of prm-PASEF and its advantages over the existing targeted and discovery methods for analyzing clinical samples. We are also highlighting the different requirements for the use of prm-PASEF on clinical samples. EXPERT OPINION prm-PASEF takes advantage of a dual ion-mobility trap enabling highly multiplexed targeted acquisition. It allows the implementation of a short chromatographic separation setup without sacrificing the number of targeted peptides. Analyzing clinical samples by prm-PASEF holds the promise to significantly improve throughput while maintaining sensitivity to detect the selected target proteins.
Collapse
Affiliation(s)
- Antoine Lesur
- Head of the Proteomics Platform, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Gunnar Dittmar
- Co-Head of the Quantitative Biology Unit, Proteomics of Cellular Signaling Research Group Luxembourg Institute of Health, Strassen, Luxembourg
| |
Collapse
|
48
|
Vadillo E, Taniguchi-Ponciano K, Lopez-Macias C, Carvente-Garcia R, Mayani H, Ferat-Osorio E, Flores-Padilla G, Torres J, Gonzalez-Bonilla CR, Majluf A, Albarran-Sanchez A, Galan JC, Peña-Martínez E, Silva-Román G, Vela-Patiño S, Ferreira-Hermosillo A, Ramirez-Renteria C, Espinoza-Sanchez NA, Pelayo-Camacho R, Bonifaz L, Arriaga-Pizano L, Mata-Lozano C, Andonegui-Elguera S, Wacher N, Blanco-Favela F, De-Lira-Barraza R, Villanueva-Compean H, Esquivel-Pineda A, Ramírez-Montes-de-Oca R, Anda-Garay C, Noyola-García M, Guizar-García L, Cerbulo-Vazquez A, Zamudio-Meza H, Marrero-Rodríguez D, Mercado M. A Shift Towards an Immature Myeloid Profile in Peripheral Blood of Critically Ill COVID-19 Patients. Arch Med Res 2021; 52:311-323. [PMID: 33248817 PMCID: PMC7670924 DOI: 10.1016/j.arcmed.2020.11.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/02/2020] [Accepted: 11/12/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND SARS-CoV-2, the etiological agent causing COVID-19, has infected more than 27 million people with over 894000 deaths worldwide since its emergence in December 2019. Factors for severe diseases, such as diabetes, hypertension, and obesity have been identified however, the precise pathogenesis is poorly understood. To understand its pathophysiology and to develop effective therapeutic strategies, it is essential to define the prevailing immune cellular subsets. METHODS We performed whole circulating immune cells scRNAseq from five critically ill COVID-19 patients, trajectory and gene ontology analysis. RESULTS Immature myeloid populations, such as promyelocytes-myelocytes, metamyelocytes, band neutrophils, monocytoid precursors, and activated monocytes predominated. The trajectory with pseudotime analysis supported the finding of immature cell states. While the gene ontology showed myeloid cell activation in immune response, DNA and RNA processing, defense response to the virus, and response to type 1 interferon. Lymphoid lineage was scarce. Expression of genes such as C/EBPβ, IRF1and FOSL2 potentially suggests the induction of trained immunity. CONCLUSIONS Our results uncover transcriptomic profiles related to immature myeloid lineages and suggest the potential induction of trained immunity.
Collapse
Affiliation(s)
- Eduardo Vadillo
- Unidad de Investigación Médica en Enfermedades Oncológicas, Hospital de Oncología, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Keiko Taniguchi-Ponciano
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Constantino Lopez-Macias
- Unidad de Investigación Médica en Inmunoquimica, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Roberto Carvente-Garcia
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México; Analitek S.A. de C.V, Ciudad de México, México
| | - Hector Mayani
- Unidad de Investigación Médica en Enfermedades Oncológicas, Hospital de Oncología, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Eduardo Ferat-Osorio
- Division de Investigacion en Salud, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Guillermo Flores-Padilla
- Servicio de Medicina Interna, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Javier Torres
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, Hospital de Pediatria, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Cesar Raul Gonzalez-Bonilla
- Coordinación de Investigación en Salud, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Abraham Majluf
- Unidad de Investigación Médica en trombosis, hemostasia y aterogenesis, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Alejandra Albarran-Sanchez
- Servicio de Medicina Interna, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Juan Carlos Galan
- Servicio de Medicina Interna, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Eduardo Peña-Martínez
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Gloria Silva-Román
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Sandra Vela-Patiño
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Aldo Ferreira-Hermosillo
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Claudia Ramirez-Renteria
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Nancy Adriana Espinoza-Sanchez
- Unidad de Investigación Médica en Enfermedades Oncológicas, Hospital de Oncología, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Rosana Pelayo-Camacho
- Centro de Investigación Biomedica de Oriente, Instituto Mexicano del Seguro Social, Puebla, México
| | - Laura Bonifaz
- Unidad de Investigación Médica en Inmunoquimica, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Lourdes Arriaga-Pizano
- Unidad de Investigación Médica en Inmunoquimica, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Carlos Mata-Lozano
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México; Analitek S.A. de C.V, Ciudad de México, México
| | - Sergio Andonegui-Elguera
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Niels Wacher
- Unidad de Investigación Médica en Epidemiologia Clinica, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Francisco Blanco-Favela
- Unidad de Investigación Médica en Inmunología, Hospital de Pediatría, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Roberto De-Lira-Barraza
- Servicio de Medicina Interna, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Humberto Villanueva-Compean
- Servicio de Medicina Interna, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Alejandra Esquivel-Pineda
- Servicio de Medicina Interna, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Rubén Ramírez-Montes-de-Oca
- Servicio de Medicina Interna, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Carlos Anda-Garay
- Servicio de Medicina Interna, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Maura Noyola-García
- Servicio de Medicina Interna, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Luis Guizar-García
- Servicio de Medicina Interna, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Arturo Cerbulo-Vazquez
- Unidad de Investigación Médica en Inmunoquimica, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Horacio Zamudio-Meza
- Unidad de Investigación Médica en Inmunoquimica, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Daniel Marrero-Rodríguez
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México.
| | - Moises Mercado
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México.
| |
Collapse
|
49
|
Cao C, Prado MA, Sun L, Rockowitz S, Sliz P, Paulo JA, Finley D, Fleming MD. Maternal Iron Deficiency Modulates Placental Transcriptome and Proteome in Mid-Gestation of Mouse Pregnancy. J Nutr 2021; 151:1073-1083. [PMID: 33693820 PMCID: PMC8112763 DOI: 10.1093/jn/nxab005] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/26/2020] [Accepted: 01/06/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Maternal iron deficiency (ID) is associated with poor pregnancy and fetal outcomes. The effect is thought to be mediated by the placenta but there is no comprehensive assessment of placental responses to maternal ID. Additionally, whether the influence of maternal ID on the placenta differs by fetal sex is unknown. OBJECTIVES To identify gene and protein signatures of ID mouse placentas at mid-gestation. A secondary objective was to profile the expression of iron genes in mouse placentas across gestation. METHODS We used a real-time PCR-based array to determine the mRNA expression of all known iron genes in mouse placentas at embryonic day (E) 12.5, E14.5, E16.5, and E19.5 (n = 3 placentas/time point). To determine the effect of maternal ID, we performed RNA sequencing and proteomics in male and female placentas from ID and iron-adequate mice at E12.5 (n = 8 dams/diet). RESULTS In female placentas, 6 genes, including transferrin receptor (Tfrc) and solute carrier family 11 member 2, were significantly changed by maternal ID. An additional 154 genes were altered in male ID placentas. A proteomic analysis quantified 7662 proteins in the placenta. Proteins translated from iron-responsive element (IRE)-containing mRNA were altered in abundance; ferritin and ferroportin 1 decreased, while TFRC increased in ID placentas. Less than 4% of the significantly altered genes in ID placentas occurred both at the transcriptional and translational levels. CONCLUSIONS Our data demonstrate that the impact of maternal ID on placental gene expression in mice is limited in scope and magnitude at mid-gestation. We provide strong evidence for IRE-based transcriptional and translational coordination of iron gene expression in the mouse placenta. Finally, we discover sexually dimorphic effects of maternal ID on placental gene expression, with more genes and pathways altered in male compared with female mouse placentas.
Collapse
Affiliation(s)
- Chang Cao
- Address correspondence to CC (e-mail: )
| | - Miguel A Prado
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Liang Sun
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, USA,The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, USA
| | - Shira Rockowitz
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, USA,The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, USA
| | - Piotr Sliz
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, USA,The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, USA,Division of Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Daniel Finley
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Mark D Fleming
- Department of Pathology, Boston Children's Hospital, Boston, MA, USA
| |
Collapse
|
50
|
Fresneda Alarcon M, McLaren Z, Wright HL. Neutrophils in the Pathogenesis of Rheumatoid Arthritis and Systemic Lupus Erythematosus: Same Foe Different M.O. Front Immunol 2021; 12:649693. [PMID: 33746988 PMCID: PMC7969658 DOI: 10.3389/fimmu.2021.649693] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 02/12/2021] [Indexed: 12/14/2022] Open
Abstract
Dysregulated neutrophil activation contributes to the pathogenesis of autoimmune diseases including rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE). Neutrophil-derived reactive oxygen species (ROS) and granule proteases are implicated in damage to and destruction of host tissues in both conditions (cartilage in RA, vascular tissue in SLE) and also in the pathogenic post-translational modification of DNA and proteins. Neutrophil-derived cytokines and chemokines regulate both the innate and adaptive immune responses in RA and SLE, and neutrophil extracellular traps (NETs) expose nuclear neoepitopes (citrullinated proteins in RA, double-stranded DNA and nuclear proteins in SLE) to the immune system, initiating the production of auto-antibodies (ACPA in RA, anti-dsDNA and anti-acetylated/methylated histones in SLE). Neutrophil apoptosis is dysregulated in both conditions: in RA, delayed apoptosis within synovial joints contributes to chronic inflammation, immune cell recruitment and prolonged release of proteolytic enzymes, whereas in SLE enhanced apoptosis leads to increased apoptotic burden associated with development of anti-nuclear auto-antibodies. An unbalanced energy metabolism in SLE and RA neutrophils contributes to the pathology of both diseases; increased hypoxia and glycolysis in RA drives neutrophil activation and NET production, whereas decreased redox capacity increases ROS-mediated damage in SLE. Neutrophil low-density granulocytes (LDGs), present in high numbers in the blood of both RA and SLE patients, have opposing phenotypes contributing to clinical manifestations of each disease. In this review we will describe the complex and contrasting phenotype of neutrophils and LDGs in RA and SLE and discuss their discrete roles in the pathogenesis of each condition. We will also review our current understanding of transcriptomic and metabolomic regulation of neutrophil phenotype in RA and SLE and discuss opportunities for therapeutic targeting of neutrophil activation in inflammatory auto-immune disease.
Collapse
Affiliation(s)
- Michele Fresneda Alarcon
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Zoe McLaren
- Liverpool University Hospitals National Health Service (NHS) Foundation Trust, Liverpool, United Kingdom
| | - Helen Louise Wright
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|