1
|
Wang J, Zhang Y, Cao C, Hua J, Xing L, Wu C. The anti-atherosclerosis effect and molecular mechanism of AMPKα1 by polarizing monocytes to an M2 phenotype via cell-intrinsic lysosomal lipolysis. Cardiovasc Pathol 2025; 78:107742. [PMID: 40354887 DOI: 10.1016/j.carpath.2025.107742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 05/09/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025] Open
Abstract
Regulating the differentiation of monocytes into M2 macrophages can promote the regression of Atherosclerosis (AS) plaque. However, the key molecules regulating the differentiation of monocytes to M2 are unknown. In this study, we reported that adenosine-activated protein kinase α1 (AMPKα1) plays an anti-AS role by polarizing monocytes to an M2 phenotype via promoting fatty acid oxidation (FAO). AMPKα1 enhances the decomposition of cholesterol esters by increasing lysosomal acid lipase expression to provide fatty acids for FAO. Furthermore, AMPKα1 can induce lysosomal biogenesis and enhance lipolysis by promoting the transcription factor EB (TFEB) expression and facilitating TFEB nuclear translocation. In conclusion, AMPKα1 enhances the decomposition of cholesterol esters by increasing lysosomal acid lipase expression to produce fatty acids, which may represent a mechanism to promote FAO and inflammatory monocytes differentiation towards M2 phenotype.
Collapse
Affiliation(s)
- Jing Wang
- Institutes of Biomedical Sciences, Shanxi University, 92 Wucheng Road, Taiyuan, 030006, Shanxi province, China.
| | - Yahui Zhang
- School of Life Science, Shanxi University, Taiyuan, 030006, China
| | - Caixing Cao
- Institutes of Biomedical Sciences, Shanxi University, 92 Wucheng Road, Taiyuan, 030006, Shanxi province, China
| | - Jiale Hua
- Institutes of Biomedical Sciences, Shanxi University, 92 Wucheng Road, Taiyuan, 030006, Shanxi province, China
| | - Li Xing
- Institutes of Biomedical Sciences, Shanxi University, 92 Wucheng Road, Taiyuan, 030006, Shanxi province, China
| | - Changxin Wu
- Institutes of Biomedical Sciences, Shanxi University, 92 Wucheng Road, Taiyuan, 030006, Shanxi province, China.
| |
Collapse
|
2
|
LeCher JC, Costa VV, Rust LN, Bassit LC, Patel D, Rezaei S, Moua J, Santos FRDS, Goncalves MR, Queroz-Junior CM, Marim FM, Zhou L, Lee S, McBrayer T, De R, Azadi N, Salman M, Zandi K, Amblard F, Burwitz B, Teixeira MM, Schinazi RF. Combating yellow fever virus with 7-deaza-7-fluoro-2'-C-methyladenosine. Antimicrob Agents Chemother 2025; 69:e0188924. [PMID: 40227063 PMCID: PMC12057363 DOI: 10.1128/aac.01889-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 03/14/2025] [Indexed: 04/15/2025] Open
Abstract
Yellow fever virus (YFV) is a deadly zoonotic flavivirus endemic in tropical/sub-tropical Africa and South America transmitted by mosquito vector (Aedes aegypti; Haemagogus leucocelaenus) to humans and non-human primates. There are no approved antiviral agents for YFV. We previously identified 7-deaza-7-fluoro-2'-C-methyladenosine (DFA) with anti-YFV activity. Interestingly, DFA exhibits pan-activity in vitro against flaviviruses, such as dengue, Japanese encephalitis, Zika, and hepatitis C. This study aimed to expand DFA's anti-flavivirus profile. DFA exhibited potent sub-micromolar anti-YFV activity in vitro against both the vaccine strain (YFV-17D) and a viscerotropic clinical YFV isolate (DakH1279) concomitantly with low cellular cytotoxicity and no notable mitochondrial toxicity. In vivo, efficacy was assessed against both YFV-17DD and a human clinical isolate in A129 and AG129 mouse flavivirus infection models, respectively. DFA significantly reduced virus replication in the livers of YFV-infected mice and the hallmarks of YFV-induced liver damage, including alanine transaminase levels and indocyanine green clearance. Collectively, this work identifies DFA as a potent YFV inhibitor and lays the groundwork for further therapeutic development as a YFV and, potentially, pan-flavivirus therapeutic.
Collapse
Affiliation(s)
- Julia C. LeCher
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Vivian Vasconcelos Costa
- Department of Morphology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, State of Minas Gerais, Brazil
| | - Lauren N. Rust
- Division of Pathobiology & Immunology, Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Leda C. Bassit
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Dharmeshkumar Patel
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Sahar Rezaei
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Justin Moua
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Felipe Rocha da Silva Santos
- Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Matheus Rodrigues Goncalves
- Department of Morphology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, State of Minas Gerais, Brazil
| | - Celso Martins Queroz-Junior
- Department of Morphology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, State of Minas Gerais, Brazil
| | - Fernanda Martins Marim
- Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Longhu Zhou
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Sujin Lee
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Tamara McBrayer
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Ramyani De
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Niloufar Azadi
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Mohammad Salman
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Keivan Zandi
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Franck Amblard
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Benjamin Burwitz
- Division of Pathobiology & Immunology, Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Mauro M. Teixeira
- Department of Morphology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, State of Minas Gerais, Brazil
| | - Raymond F. Schinazi
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| |
Collapse
|
3
|
Kuchenreuther I, Clausen FN, Mazurie J, Paul S, Czubayko F, Mittelstädt A, Koch AK, Karabiber A, Hansen FJ, Arnold LS, Weisel N, Merkel S, Brunner M, Krautz C, Vera J, Grützmann R, Weber GF, David P. Increased Herpesvirus Entry Mediator Expression on Circulating Monocytes and Subsets Predicts Poor Outcomes in Pancreatic Ductal Adenocarcinoma Patients. Int J Mol Sci 2025; 26:2875. [PMID: 40243455 PMCID: PMC11988668 DOI: 10.3390/ijms26072875] [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: 01/16/2025] [Revised: 03/11/2025] [Accepted: 03/18/2025] [Indexed: 04/18/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is aggressive, with a 5-year survival rate of only 12.8%, and its increasing incidence in Western countries highlights the urgent need for better early-stage detection and treatment methods. Early diagnosis significantly improves the chances of survival, but non-specific symptoms and undetectable precursor lesions pose a major challenge. To date, there are no reliable screening tools to detect PDAC at an early stage. Herpesvirus entry mediator (HVEM) has already been proposed as a prognostic marker in numerous cancer types. Therefore, we investigated the role of HVEM in PDAC. Flow cytometry was used to analyze HVEM expression in immune cells and its inhibitory receptors (CD160 and BTLA) on T-cells, as well as its subsets in the peripheral blood of 57 diagnosed PDAC patients and 17 clinical controls. In addition, survival analyses were performed within the PDAC cohort, changes in HVEM expression were analyzed in relation to clinicopathological parameters, and a correlation analysis between HVEM expression and cytokine levels of IL-6 and IL-10 was conducted. Furthermore, HVEM expression on monocytes and their subsets was evaluated as a potential prognostic marker and compared with the prognostic utility of CA19-9. We found that HVEM expression is significantly elevated on immune cells, particularly on monocytes (p < 0.0001) and their subsets, in PDAC patients, and is associated with reduced survival (p = 0.0067) and clinicopathological features such as perineural, lymphovascular, and vascular invasion. Moreover, HVEM-expressing monocytes demonstrated superior predictive value compared to CA19-9, highlighting their potential as part of a combined screening tool for PDAC. In conclusion, HVEM on monocytes could serve as a novel prognostic marker for PDAC.
Collapse
MESH Headings
- Humans
- Receptors, Tumor Necrosis Factor, Member 14/metabolism
- Receptors, Tumor Necrosis Factor, Member 14/blood
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/blood
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/mortality
- Carcinoma, Pancreatic Ductal/diagnosis
- Female
- Male
- Middle Aged
- Monocytes/metabolism
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/blood
- Pancreatic Neoplasms/mortality
- Prognosis
- Aged
- Biomarkers, Tumor/blood
- Biomarkers, Tumor/metabolism
- Antigens, CD/metabolism
- Receptors, Immunologic/metabolism
- Interleukin-10
- Adult
- Interleukin-6
- GPI-Linked Proteins
Collapse
Affiliation(s)
- Isabelle Kuchenreuther
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
- Faculty of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Finn-Niklas Clausen
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
- Faculty of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Johanne Mazurie
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
- Faculty of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Sushmita Paul
- Department of Dermatology, University Hospital Erlangen, 91054 Erlangen, Germany; (S.P.); (J.V.)
| | - Franziska Czubayko
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
| | - Anke Mittelstädt
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
| | - Ann-Kathrin Koch
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
| | - Alara Karabiber
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
| | - Frederik J. Hansen
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
| | - Lisa-Sophie Arnold
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
- Faculty of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Nadine Weisel
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
| | - Susanne Merkel
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
| | - Maximilian Brunner
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
| | - Christian Krautz
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
| | - Julio Vera
- Department of Dermatology, University Hospital Erlangen, 91054 Erlangen, Germany; (S.P.); (J.V.)
| | - Robert Grützmann
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
- Faculty of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg and University Hospital Erlangen, 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91052 Erlangen, Germany
| | - Georg F. Weber
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
- Faculty of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg and University Hospital Erlangen, 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91052 Erlangen, Germany
| | - Paul David
- Department of Surgery, University Hospital Erlangen, 91054 Erlangen, Germany; (I.K.); (F.-N.C.); (J.M.); (F.C.); (A.M.); (A.-K.K.); (A.K.); (F.J.H.); (L.-S.A.); (N.W.); (S.M.); (M.B.); (C.K.); (R.G.); (P.D.)
- Faculty of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| |
Collapse
|
4
|
Lucotti S, Ogitani Y, Kenific CM, Geri J, Kim YH, Gu J, Balaji U, Bojmar L, Shaashua L, Song Y, Cioffi M, Lauritzen P, Joseph OM, Asao T, Grandgenett PM, Hollingsworth MA, Peralta C, Pagano AE, Molina H, Lengel HB, Dunne EG, Jing X, Schmitter M, Borriello L, Miller T, Zhang H, Romin Y, Manova K, Paul D, Remmel HL, O'Reilly EM, Jarnagin WR, Kelsen D, Castellino SM, Giulino-Roth L, Jones DR, Condeelis JS, Pascual V, Bussel JB, Boudreau N, Matei I, Entenberg D, Bromberg JF, Simeone DM, Lyden D. Extracellular vesicles from the lung pro-thrombotic niche drive cancer-associated thrombosis and metastasis via integrin beta 2. Cell 2025; 188:1642-1661.e24. [PMID: 39938515 DOI: 10.1016/j.cell.2025.01.025] [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: 01/30/2024] [Revised: 08/08/2024] [Accepted: 01/15/2025] [Indexed: 02/14/2025]
Abstract
Cancer is a systemic disease with complications beyond the primary tumor site. Among them, thrombosis is the second leading cause of death in patients with certain cancers (e.g., pancreatic ductal adenocarcinoma [PDAC]) and advanced-stage disease. Here, we demonstrate that pro-thrombotic small extracellular vesicles (sEVs) are secreted by C-X-C motif chemokine 13 (CXCL13)-reprogrammed interstitial macrophages in the non-metastatic lung microenvironment of multiple cancers, a niche that we define as the pro-thrombotic niche (PTN). These sEVs package clustered integrin β2 that dimerizes with integrin αX and interacts with platelet-bound glycoprotein (GP)Ib to induce platelet aggregation. Blocking integrin β2 decreases both sEV-induced thrombosis and lung metastasis. Importantly, sEV-β2 levels are elevated in the plasma of PDAC patients prior to thrombotic events compared with patients with no history of thrombosis. We show that lung PTN establishment is a systemic consequence of cancer progression and identify sEV-β2 as a prognostic biomarker of thrombosis risk as well as a target to prevent thrombosis and metastasis.
Collapse
Affiliation(s)
- Serena Lucotti
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Drukier Institute for Children's Health and Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA.
| | - Yusuke Ogitani
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Drukier Institute for Children's Health and Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Candia M Kenific
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Drukier Institute for Children's Health and Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Jacob Geri
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Young Hun Kim
- Molecular Cytology Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jinghua Gu
- Drukier Institute for Children's Health and Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Uthra Balaji
- Drukier Institute for Children's Health and Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Linda Bojmar
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Drukier Institute for Children's Health and Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA; Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Lee Shaashua
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Drukier Institute for Children's Health and Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Yi Song
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michele Cioffi
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Drukier Institute for Children's Health and Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Pernille Lauritzen
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Drukier Institute for Children's Health and Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Oveen M Joseph
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Drukier Institute for Children's Health and Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Tetsuhiko Asao
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Drukier Institute for Children's Health and Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA; Thoracic Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Respiratory Medicine, Juntendo University, Tokyo, Japan
| | - Paul M Grandgenett
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | - Michael A Hollingsworth
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | | | - Alexandra E Pagano
- Proteomics Resource Center, The Rockefeller University, New York, NY, USA
| | - Henrik Molina
- Proteomics Resource Center, The Rockefeller University, New York, NY, USA
| | - Harry B Lengel
- Thoracic Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elizabeth G Dunne
- Thoracic Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Xiaohong Jing
- Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Madeleine Schmitter
- Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Lucia Borriello
- Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA; Fox Chase Cancer Center, Cancer Signaling and Microenvironment Program, Philadelphia, PA, USA
| | - Thomas Miller
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Drukier Institute for Children's Health and Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Haiying Zhang
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Drukier Institute for Children's Health and Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Yevgeniy Romin
- Molecular Cytology Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Katia Manova
- Molecular Cytology Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Doru Paul
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - H Lawrence Remmel
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Drukier Institute for Children's Health and Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA; Atossa Therapeutics, Inc., Seattle, WA, USA; Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Eileen M O'Reilly
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - William R Jarnagin
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David Kelsen
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sharon M Castellino
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA; Aflac Cancer & Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Lisa Giulino-Roth
- Department of Pediatrics, Division of Hematology/Oncology, Weill Cornell Medicine, New York, NY, USA
| | - David R Jones
- Thoracic Surgery Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John S Condeelis
- Department of Surgery, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA; Integrated Imaging Program for Cancer Research, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA; Montefiore Einstein Comprehensive Cancer Center, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA; Gruss-Lipper Biophotonics Center, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA; Department of Cell Biology, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA; Cancer Dormancy Institute, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Virginia Pascual
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Drukier Institute for Children's Health and Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - James B Bussel
- Drukier Institute for Children's Health and Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA; Department of Pediatrics, Division of Hematology/Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Nancy Boudreau
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Drukier Institute for Children's Health and Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Irina Matei
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Drukier Institute for Children's Health and Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - David Entenberg
- Integrated Imaging Program for Cancer Research, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA; Montefiore Einstein Comprehensive Cancer Center, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA; Gruss-Lipper Biophotonics Center, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA; Department of Cell Biology, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA; Cancer Dormancy Institute, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA; Department of Pathology, Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Jacqueline F Bromberg
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| | - Diane M Simeone
- Department of Surgery, UC San Diego Health, San Diego, CA, USA; Moores Cancer Center, UC San Diego Health, San Diego, CA, USA.
| | - David Lyden
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Drukier Institute for Children's Health and Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
5
|
Lin MH, Maniam P, Li D, Tang B, Bishop CR, Suhrbier A, Earl LW, Tayyar Y, McMillan NA, Li L, Harrich D. Harnessing defective interfering particles and lipid nanoparticles for effective delivery of an anti-dengue virus RNA therapy. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102424. [PMID: 39817192 PMCID: PMC11733052 DOI: 10.1016/j.omtn.2024.102424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 12/10/2024] [Indexed: 01/18/2025]
Abstract
Currently, no approved antiviral drugs target dengue virus (DENV) infection, leaving treatment reliant on supportive care. DENV vaccine efficacy varies depending on the vaccine type, the circulating serotype, and vaccine coverage. We investigated defective interfering particles (DIPs) and lipid nanoparticles (LNPs) to deliver DI290, an anti-DENV DI RNA. Both DIPs and DI290-loaded LNPs (LNP-290) effectively suppressed DENV infection in human primary monocyte-derived macrophages (MDMs), THP-1 macrophages, and fibroblasts-natural DENV targets. Inhibiting interferon (IFN) signaling with a Janus kinase 1/2 inhibitor or an IFN-α/β receptor 1 (IFNAR1)-binding antibody blocked DIP and LNP-290 antiviral activity. LNP-290 demonstrated a greater than log10 inhibition of DENV viral loads in IFNAR-deficient (Ifnar -/- ) and IFN regulatory factor (IRF) 3 and 7 double knockout (Irf3/7 -/- ) mice. Pathway analysis of RNA sequencing data from LNP-treated C57BL/6J mice, Ifnar -/- mice, and human MDMs treated with LNPs or DENV DIPs indicated DI290 treatment enhanced IFN responses, suggesting IFN-λ and IFN-γ provided antiviral activity when IFN-α/β responses were diminished. While viral interference by DI290 is possible, results did not support RNA replication competition as an inhibition mechanism. These findings suggest that DI290 may be a promising DENV therapeutic by activating the innate immune system.
Collapse
Affiliation(s)
- Min-Hsuan Lin
- Program of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Pramila Maniam
- Program of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Dongsheng Li
- Program of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Bing Tang
- Program of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Cameron R. Bishop
- Program of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Andreas Suhrbier
- Program of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
- Global Virus Network (GVN) Center of Excellence, Australian Infectious Disease Research Centre, Brisbane, QLD 4072, Australia
| | - Lucy Wales- Earl
- Menzies Health Institute Queensland and School of Pharmacy and Medical Science, Griffith University, Gold Coast, QLD 4222, Australia
| | - Yaman Tayyar
- Menzies Health Institute Queensland and School of Pharmacy and Medical Science, Griffith University, Gold Coast, QLD 4222, Australia
- Prorenata Biotech, Molendinar, QLD 4214, Australia
| | - Nigel A.J. McMillan
- Menzies Health Institute Queensland and School of Pharmacy and Medical Science, Griffith University, Gold Coast, QLD 4222, Australia
| | - Li Li
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, St Lucia, QLD 4072, Australia
| | - David Harrich
- Program of Infection and Inflammation, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
- Global Virus Network (GVN) Center of Excellence, Australian Infectious Disease Research Centre, Brisbane, QLD 4072, Australia
| |
Collapse
|
6
|
Antonsen KW, Jensen AG, Sorensen BS, Etzerodt A, Moestrup SK, Møller HJ. In vitro ovarian tumor-conditioned CD163+ human macrophages retain phagocytic response to CD47 blockade. Cell Immunol 2025; 409-410:104932. [PMID: 39985839 DOI: 10.1016/j.cellimm.2025.104932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 02/02/2025] [Accepted: 02/13/2025] [Indexed: 02/24/2025]
Abstract
INTRODUCTION CD163-expressing macrophages are abundant in ovarian cancer where they accelerate tumor growth and metastasis. CD47 blockade is a novel immunotherapy aiming to activate macrophage phagocytosis of tumor cells, but it is currently unknown if the tumor-associated macrophages expressing CD163 respond poorly to CD47 blockade. METHODS Human monocyte-derived macrophages were exposed to tumor-conditioned medium from A2780 ovarian cancer cells during differentiation. Effects on gene expression, membrane protein levels, release of soluble proteins and macrophage phagocytosis of A2780 cells in response to CD47 blockade were measured and compared to control macrophages. RESULTS Tumor cell conditioning induced macrophage expression of CD163 on both the mRNA and protein level. Furthermore, tumor conditioning simultaneously increased protein expression of the phenotype markers CD206 and CD80, and the phagocytosis checkpoint LILRB1. However, tumor conditioning did not reduce phagocytic capacity, as CD47 blockade induced macrophage phagocytosis of A2780 cells to similar degrees in both control and tumor cell-conditioned macrophages. DISCUSSION In vitro tumor conditioning did not reduce the phagocytic response to CD47 blockade, suggesting that induction of a macrophage phenotype with increased expression of CD163 does not directly limit the capacity for phagocytosis of tumor cells. In conclusion, these findings suggest that CD163+ macrophages remain responsive to CD47 blockade, highlighting their potential as targets for immunotherapy in ovarian cancer.
Collapse
MESH Headings
- Humans
- Female
- Ovarian Neoplasms/immunology
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/pathology
- Phagocytosis/immunology
- CD163 Antigen
- Receptors, Cell Surface/metabolism
- Receptors, Cell Surface/immunology
- CD47 Antigen/immunology
- CD47 Antigen/antagonists & inhibitors
- CD47 Antigen/metabolism
- Antigens, CD/metabolism
- Antigens, CD/immunology
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, Differentiation, Myelomonocytic/immunology
- Macrophages/immunology
- Macrophages/metabolism
- Cell Line, Tumor
- Receptors, Immunologic/metabolism
- Culture Media, Conditioned/pharmacology
- Tumor-Associated Macrophages/immunology
- Tumor-Associated Macrophages/metabolism
- Immunotherapy/methods
- Cell Differentiation
Collapse
Affiliation(s)
- Kristian W Antonsen
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| | - Anne G Jensen
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Boe S Sorensen
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Anders Etzerodt
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Holger J Møller
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
7
|
Ishihara N, Koma YI, Omori M, Komatsu S, Torigoe R, Yokoo H, Nakanishi T, Yamanaka K, Azumi Y, Tsukamoto S, Kodama T, Nishio M, Shigeoka M, Yokozaki H, Fukumoto T. Chemokine (C-C Motif) Ligand 2/CCR2/Extracellular Signal-Regulated Kinase Signal Induced through Cancer Cell-Macrophage Interaction Contributes to Hepatocellular Carcinoma Progression. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:589-608. [PMID: 39756577 DOI: 10.1016/j.ajpath.2024.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/23/2024] [Accepted: 12/27/2024] [Indexed: 01/07/2025]
Abstract
Tumor-infiltrating macrophages, known as tumor-associated macrophages, play a crucial role in the tumor microenvironment. Herein, immunohistochemistry revealed that intratumoral CD68-positive macrophages are associated with poor prognosis and clinicopathologic factors in patients with hepatocellular carcinoma (HCC). Subsequently, an indirect co-culture system involving HCC cells and peripheral blood-derived macrophages was developed. cDNA microarray analysis revealed that chemokine (C-C motif) ligand 2 (CCL2) was highly expressed in HCC cells co-cultured with macrophages. CCL2 neutralization suppressed proliferation, migration, and phosphorylation of extracellular signal-regulated kinase (Erk) in HCC cells and macrophages enhanced through co-culture. In contrast, recombinant human CCL2 (rhCCL2) addition facilitated these malignant phenotypes and increased Erk phosphorylation levels in HCC cells and macrophages. The primary CCL2 receptor, CCR2, was expressed in HCC cells and macrophages and was up-regulated in co-cultured HCC cells. CCR2 inhibition suppressed malignant phenotypes and reduced phosphorylated levels of Erk enhanced by rhCCL2. Additionally, the inhibition of Erk signal suppressed rhCCL2-enhanced malignant phenotypes. Moreover, serum CCL2 levels were higher in patients with HCC than those in healthy donors. On the basis of immunohistochemistry, CCL2-positive cases with high CCR2 expression and phosphorylated Erk-positive cases exhibited poor survival outcomes. Therefore, CCL2 up-regulation through interactions between HCC cells and macrophages contributed to HCC progression, making the CCL2/CCR2/Erk signal a potential target for HCC treatment.
Collapse
Affiliation(s)
- Nobuaki Ishihara
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan; Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yu-Ichiro Koma
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - Masaki Omori
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan; Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shohei Komatsu
- Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Rikuya Torigoe
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan; Division of Gastro-Intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiroki Yokoo
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan; Division of Gastro-Intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takashi Nakanishi
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan; Division of Gastro-Intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Keitaro Yamanaka
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan; Division of Obstetrics and Gynecology, Department of Surgery Related, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuki Azumi
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan; Division of Gastro-Intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shuichi Tsukamoto
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takayuki Kodama
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Mari Nishio
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Manabu Shigeoka
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiroshi Yokozaki
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takumi Fukumoto
- Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
8
|
Landwehr KR, Larcombe AN. Comment on Karthikeyan et al. Concordance between In Vitro and In Vivo Relative Toxic Potencies of Diesel Exhaust Particles from Different Biodiesel Blends. Toxics 2024, 12, 290. TOXICS 2025; 13:174. [PMID: 40137501 PMCID: PMC11945800 DOI: 10.3390/toxics13030174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 01/04/2025] [Accepted: 02/11/2025] [Indexed: 03/29/2025]
Abstract
Biodiesel exhaust toxicology is a difficult field of study, for which there is a paucity of literature, despite decades of research into the subject [...].
Collapse
Affiliation(s)
- Katherine R. Landwehr
- Occupation, Environment and Safety, School of Population Health, Curtin University, Perth, WA 6845, Australia;
- Respiratory Environmental Health, Wal-yan Respiratory Research Centre, The Kids Research Institute Australia, Perth Children’s Hospital, Nedlands, Perth, WA 6009, Australia
| | - Alexander N. Larcombe
- Occupation, Environment and Safety, School of Population Health, Curtin University, Perth, WA 6845, Australia;
- Respiratory Environmental Health, Wal-yan Respiratory Research Centre, The Kids Research Institute Australia, Perth Children’s Hospital, Nedlands, Perth, WA 6009, Australia
- School of Human Sciences, University of Western Australia, Crawley, WA 6009, Australia
| |
Collapse
|
9
|
Gohara Y, Kinoshita R, Tomonobu N, Jiang F, Matsunaga Y, Hashimoto Y, Honjo T, Yamamoto KI, Murata H, Ochi T, Komalasari NLGY, Yamauchi A, Kuribayashi F, Sakaguchi Y, Futami J, Inoue Y, Kondo E, Toyooka S, Morizane S, Ishiko A, Morita S, Sagayama K, Nakao K, Sakaguchi M. An S100A8/A9 Neutralizing Antibody Potently Ameliorates Contact Hypersensitivity and Atopic Dermatitis Symptoms. J Invest Dermatol 2025:S0022-202X(25)00029-6. [PMID: 39848567 DOI: 10.1016/j.jid.2025.01.007] [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: 04/03/2024] [Revised: 12/07/2024] [Accepted: 01/01/2025] [Indexed: 01/25/2025]
Abstract
Contact hypersensitivity and atopic dermatitis are pervasive inflammatory skin diseases with similar symptoms, and their global prevalence is steadily increasing. Many compounds and biotics have been developed to target molecules critical to the etiology or pathogenesis of contact hypersensitivity and atopic dermatitis. However, these molecules are sometimes ineffective or lose their potency during the therapeutic course. Therefore, innovative medicines are still needed for the treatment of intractable cases. We focused on S100A8/A9, a heterodimer complex of S100A8 and S100A9 that is abundant in the extracellular milieu of inflammatory skin lesions. Although S100A8/A9 is primarily recognized as a diagnostic marker protein, we have previously shown that it also plays a crucial role in contact hypersensitivity and atopic dermatitis progression. This insight inspired us to develop its inhibitory antibody, leading to the ground-breaking Ab45. In this study, we demonstrated that Ab45 effectively prevented disease symptoms in various models and that its disease-ameliorating activity likely involved the downregulation of several disease-relevant molecules, including Il-23a, Il-36g, S100a8, and S100a9. We also created a humanized version of Ab45, HuAb45, which exhibited similar effectiveness. These antibodies show great promise for the treatment of contact hypersensitivity and atopic dermatitis and possibly for other inflammatory skin diseases.
Collapse
Affiliation(s)
- Yuma Gohara
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Rie Kinoshita
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Nahoko Tomonobu
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Fan Jiang
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yukiko Matsunaga
- Department of Dermatology, Toho University Graduate School of Medicine, Tokyo, Japan
| | - Yuki Hashimoto
- Department of Dermatology, Toho University Graduate School of Medicine, Tokyo, Japan
| | - Tomoko Honjo
- Department of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, Japan
| | - Ken-Ichi Yamamoto
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hitoshi Murata
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Toshiki Ochi
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan; Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | | | - Akira Yamauchi
- Department of Biochemistry, Kawasaki Medical School, Kurashiki, Japan
| | | | | | - Junichiro Futami
- Department of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, Japan
| | - Yusuke Inoue
- Division of Molecular Science, Faculty of Science and Technology, Gunma University, Kiryu, Japan
| | - Eisaku Kondo
- Division of Tumor Pathology, Near-Infrared Photo-Immunotherapy Research Institute, Kansai Medical University, Osaka, Japan
| | - Shinichi Toyooka
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shin Morizane
- Department of Dermatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Akira Ishiko
- Department of Dermatology, Toho University Graduate School of Medicine, Tokyo, Japan
| | - Shigeru Morita
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama, Japan
| | - Kazumi Sagayama
- Organization for Research and Innovation Strategy, Okayama University, Okayama, Japan
| | | | - Masakiyo Sakaguchi
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| |
Collapse
|
10
|
Zhang H, Cao F, Zhou Y, Wu B, Li C. Peripheral Immune Cells Contribute to the Pathogenesis of Alzheimer's Disease. Mol Neurobiol 2025; 62:264-270. [PMID: 38842674 DOI: 10.1007/s12035-024-04266-6] [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: 12/05/2023] [Accepted: 05/26/2024] [Indexed: 06/07/2024]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder with progressive memory and cognitive loss. Neuroinflammation is a central mechanism involved in the progression of AD. With the disruption of the blood-brain barrier (BBB), peripheral immune cells and inflammatory molecules enter into AD brain. However, the exact relationship between peripheral immune cells and AD remains unknown due to various challenges. This study aimed to investigate the potential causal association between peripheral immune cells and AD by using a two-sample Mendelian randomization (TSMR) analysis. We conducted a TSMR to decipher the causal relationship between AD and 731 types of peripheral immune cell parameters from the TBNK, regulatory T cell (Treg), myeloid cell, monocyte, maturation stages of T cell, dendritic cell (DC), and B cell panels. Various analytical methods were employed, including inverse variance weighting (IVW), MR Egger, and weighted median methods. The Cochran's Q statistic, MR-Egger intercept, and MR-PRESSO tests were used to verify the heterogeneity and horizontal pleiotropy of the results. To further verify our results, we also conducted a replication analysis. The analysis identified CD33 on CD14 + monocyte (OR = 1.03; 95% CI, 1.01-1.04; p = 1.14E-04; adjust-p = 0.042) had an increased risk association for AD, which was verified by the replication study. CD33 on CD33dim HLA DR + CD11b- cell (OR = 1.02; 95% CI, 1.01-1.04; p = 2.87E-04; adjust-p = 0.035) had an increased risk association for AD, while secreting CD4 regulatory T cell %CD4 regulatory T cell (OR = 0.97; 95% CI, 0.96-0.99; p = 1.90E-04; adjust-p = 0.046) and CD25 on switched memory B cell (OR = 0.95; 95% CI, 0.92-0.98; p = 2.87E-04; adjust-p = 0.042) were discovered to be related to a lower risk of AD. However, the causal effect of these three immune cells on AD was insufficiently validated in the replication analysis. The MR analysis suggests a potential causal relationship between peripheral immune cells and the risk of AD. Further extensive research is needed on the specific role of peripheral immune cells in AD.
Collapse
Affiliation(s)
- Houwen Zhang
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, 310014, Zhejiang, China
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fangzheng Cao
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, 310014, Zhejiang, China
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yu Zhou
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China
| | - Bin Wu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chunrong Li
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, 310014, Zhejiang, China.
| |
Collapse
|
11
|
Meskini M, Amanzadeh A, Salehi F, Bouzari S, Karimipoor M, Fuso A, Fateh A, Siadat SD. A protocol to isolate and characterize pure monocytes and generate monocyte-derived dendritic cells through FBS-Coated flasks. Sci Rep 2024; 14:23956. [PMID: 39397067 PMCID: PMC11471755 DOI: 10.1038/s41598-024-75376-3] [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: 02/21/2024] [Accepted: 10/04/2024] [Indexed: 10/15/2024] Open
Abstract
This study explores methods to isolate high-pure monocytes and optimize the best growth factor concentration to generate monocytes-derived dendritic cells (mo-DCs), subset DC1, which is crucial in immune responses. Three protocols for monocyte isolation from peripheral blood mononuclear cells (PBMCs) were evaluated: three-hour incubation on FBS-coated flasks; an overnight incubation on FBS-coated flasks; and Magnetic Activated Cell Sorting (MACS). Additionally, five different concentrations of human recombinant granulocyte-macrophage colony-stimulating factor (hrGM-CSF) and human recombinant interleukin-4 (hrIL-4) were compared. We used Flow cytometry to assess the isolation, purification, and generation of pure monocytes characterized as CD14+, and expression of mo-DC classical markers (HLA-DR, CD80, CD83, and CD86). The obtained results show that monocytes isolated with the second method (overnight incubation) had the highest purity (P < 0.0001) but the lowest yield (P > 0.05), balancing purity and cost-effectiveness. A combination of hrGM-CSF and hrIL-4 at 400 U/mL produced the most favorable outcomes, leading to the highest rate of mo-DC generation (P < 0.05). Notably, this concentration resulted in increasing expression of HLA-DR, CD80, and CD86 surface markers in the generated DCs (P < 0.0001), with no changes in CD83 expression levels. In conclusion, this study offers valuable insights into selecting the optimal approach for monocyte isolation and mo-DC generation in various research contexts, providing a foundation for more effective immunological studies.
Collapse
Affiliation(s)
- Maryam Meskini
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
- Student Research Committee, Pasteur Institute of Iran, Tehran, Iran
| | - Amir Amanzadeh
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran
| | - Fahimeh Salehi
- Institute of Biochemistry and Biophysics, Department of Biochemistry, University of Tehran, Tehran, Iran
| | - Saeid Bouzari
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| | - Morteza Karimipoor
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
- Department of Molecular Medicine, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Andrea Fuso
- Department of Experimental Medicine, Sapienza University of Rome, Rome, 00161, Italy
| | - Abolfazl Fateh
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
12
|
Herb M, Schatz V, Hadrian K, Hos D, Holoborodko B, Jantsch J, Brigo N. Macrophage variants in laboratory research: most are well done, but some are RAW. Front Cell Infect Microbiol 2024; 14:1457323. [PMID: 39445217 PMCID: PMC11496307 DOI: 10.3389/fcimb.2024.1457323] [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: 06/30/2024] [Accepted: 09/06/2024] [Indexed: 10/25/2024] Open
Abstract
Macrophages play a pivotal role in the innate immune response. While their most characteristic function is phagocytosis, it is important not to solely characterize macrophages by this activity. Their crucial roles in body development, homeostasis, repair, and immune responses against pathogens necessitate a broader understanding. Macrophages exhibit remarkable plasticity, allowing them to modify their functional characteristics in response to the tissue microenvironment (tissue type, presence of pathogens or inflammation, and specific signals from neighboring cells) swiftly. While there is no single defined "macrophage" entity, there is a diverse array of macrophage types because macrophage ontogeny involves the differentiation of progenitor cells into tissue-resident macrophages, as well as the recruitment and differentiation of circulating monocytes in response to tissue-specific cues. In addition, macrophages continuously sense and respond to environmental cues and tissue conditions, adjusting their functional and metabolic states accordingly. Consequently, it is of paramount importance to comprehend the heterogeneous origins and functions of macrophages employed in in vitro studies, as each available in vitro macrophage model is associated with specific sets of strengths and limitations. This review centers its attention on a comprehensive comparison between immortalized mouse macrophage cell lines and primary mouse macrophages. It provides a detailed analysis of the strengths and weaknesses inherent in these in vitro models. Finally, it explores the subtle distinctions between diverse macrophage cell lines, offering insights into numerous factors beyond the model type that can profoundly influence macrophage function.
Collapse
Affiliation(s)
- Marc Herb
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Valentin Schatz
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Karina Hadrian
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Deniz Hos
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Bohdan Holoborodko
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg and University of Regensburg, Regensburg, Germany
| | - Jonathan Jantsch
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Natascha Brigo
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
13
|
Liu X, Ren Z, Tan C, Núñez-Santana FL, Kelly ME, Yan Y, Sun H, Abdala-Valencia H, Yang W, Wu Q, Toyoda T, Milisav M, Casalino-Matsuda SM, Lecuona E, Cerier EJ, Heung LJ, Abazeed ME, Perlman H, Gao R, Chandel NS, Budinger GS, Bharat A. Inducible CCR2+ nonclassical monocytes mediate the regression of cancer metastasis. J Clin Invest 2024; 134:e179527. [PMID: 39545417 PMCID: PMC11563681 DOI: 10.1172/jci179527] [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: 01/19/2024] [Accepted: 09/26/2024] [Indexed: 11/17/2024] Open
Abstract
A major limitation of immunotherapy is the development of resistance resulting from cancer-mediated inhibition of host lymphocytes. Cancer cells release CCL2 to recruit classical monocytes expressing its receptor CCR2 for the promotion of metastasis and resistance to immunosurveillance. In the circulation, some CCR2-expressing classical monocytes lose CCR2 and differentiate into intravascular nonclassical monocytes that have anticancer properties but are unable to access extravascular tumor sites. We found that in mice and humans, an ontogenetically distinct subset of naturally underrepresented CCR2-expressing nonclassical monocytes was expanded during inflammatory states such as organ transplant and COVID-19 infection. These cells could be induced during health by treatment of classical monocytes with small-molecule activators of NOD2. The presence of CCR2 enabled these inducible nonclassical monocytes to infiltrate both intra- and extravascular metastatic sites of melanoma, lung, breast, and colon cancer in murine models, and they reversed the increased susceptibility of Nod2-/- mutant mice to cancer metastasis. Within the tumor colonies, CCR2+ nonclassical monocytes secreted CCL6 to recruit NK cells that mediated tumor regression, independent of T and B lymphocytes. Hence, pharmacological induction of CCR2+ nonclassical monocytes might be useful for immunotherapy-resistant cancers.
Collapse
Affiliation(s)
- Xianpeng Liu
- Division of Thoracic Surgery/Canning Thoracic Institute, Feinberg School of Medicine, Northwestern University/Northwestern Medicine, Chicago, Illinois, USA
| | | | - Can Tan
- Division of Cardiology, Department of Medicine, and
| | - Félix L. Núñez-Santana
- Division of Thoracic Surgery/Canning Thoracic Institute, Feinberg School of Medicine, Northwestern University/Northwestern Medicine, Chicago, Illinois, USA
| | - Megan E. Kelly
- Division of Thoracic Surgery/Canning Thoracic Institute, Feinberg School of Medicine, Northwestern University/Northwestern Medicine, Chicago, Illinois, USA
| | - Yuanqing Yan
- Division of Thoracic Surgery/Canning Thoracic Institute, Feinberg School of Medicine, Northwestern University/Northwestern Medicine, Chicago, Illinois, USA
| | - Haiying Sun
- Division of Thoracic Surgery/Canning Thoracic Institute, Feinberg School of Medicine, Northwestern University/Northwestern Medicine, Chicago, Illinois, USA
| | - Hiam Abdala-Valencia
- Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Wenbin Yang
- Division of Thoracic Surgery/Canning Thoracic Institute, Feinberg School of Medicine, Northwestern University/Northwestern Medicine, Chicago, Illinois, USA
| | - Qiang Wu
- Division of Thoracic Surgery/Canning Thoracic Institute, Feinberg School of Medicine, Northwestern University/Northwestern Medicine, Chicago, Illinois, USA
| | - Takahide Toyoda
- Division of Thoracic Surgery/Canning Thoracic Institute, Feinberg School of Medicine, Northwestern University/Northwestern Medicine, Chicago, Illinois, USA
| | - Marija Milisav
- Division of Thoracic Surgery/Canning Thoracic Institute, Feinberg School of Medicine, Northwestern University/Northwestern Medicine, Chicago, Illinois, USA
| | - S. Marina Casalino-Matsuda
- Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Emilia Lecuona
- Division of Thoracic Surgery/Canning Thoracic Institute, Feinberg School of Medicine, Northwestern University/Northwestern Medicine, Chicago, Illinois, USA
| | - Emily Jeong Cerier
- Division of Thoracic Surgery/Canning Thoracic Institute, Feinberg School of Medicine, Northwestern University/Northwestern Medicine, Chicago, Illinois, USA
| | - Lena J. Heung
- Division of Infectious Diseases, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | | | | | - Ruli Gao
- Department of Biochemistry, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Navdeep S. Chandel
- Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - G.R. Scott Budinger
- Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ankit Bharat
- Division of Thoracic Surgery/Canning Thoracic Institute, Feinberg School of Medicine, Northwestern University/Northwestern Medicine, Chicago, Illinois, USA
- Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
14
|
Jha A, Moore E. YIGSR, A Laminin-Derived Peptide, Dictates a Concentration-Dependent Impact on Macrophage Phenotype Response. Cell Mol Bioeng 2024; 17:423-440. [PMID: 39513005 PMCID: PMC11538123 DOI: 10.1007/s12195-024-00810-5] [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: 02/09/2024] [Accepted: 06/18/2024] [Indexed: 11/15/2024] Open
Abstract
Purpose Macrophage immune cells play crucial roles in the inflammatory (M1) and regenerative (M2) processes. The extracellular matrix (ECM) composition, including presentation of embedded ligands, governs macrophage function. Laminin concentration is abundant in the basement membrane and is dependent on pathological state: reduced in inflammation and increased during regeneration. Distinct laminin ligands, such as IKVAV and YIGSR, have disparate roles in dictating cell function. For example, IKVAV, derived from the alpha chain of laminin, promotes angiogenesis and metastasis of cancer cells whereas YIGSR, beta chain derived, impedes angiogenesis and tumor progression. Previous work has demonstrated IKVAV's inflammation inhibiting properties in macrophages. Given the divergent role of IKVAV and YIGSR in interacting with cells through varied integrin receptors, we ask: what role does laminin derived peptide YIGSR play in governing macrophage function? Methods We quantified the influence of YIGSR on macrophage phenotype in 2D and 3D via immunostaining assessments for M1 marker inducible nitric oxide synthase (iNOS) and M2 marker Arginase-1 (Arg-1). We also analysed the secretome of human and murine macrophage response to YIGSR via a Luminex bead assay. Results YIGSR impact on macrophage phenotype occurs in a concentration-dependent manner. At lower concentrations of YIGSR, macrophage inflammation was increased whereas, at higher concentrations of YIGSR the opposite effect was seen within the same time frame. Secretomic assessments also demonstrate that pro-inflammatory chemokines and cytokines were increased at low YIGSR concentrations in M0, M1, M2 macrophages while pro-inflammatory secretion was reduced at higher concentrations. Conclusions YIGSR can be used as a tool to modulate macrophage inflammatory state within M1 and M2 phenotypes depending on the concentration of peptide. YIGSR's impact on macrophage function can be leveraged for the development of immunoengineering strategies in regenerative medicine and cancer therapy. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-024-00810-5.
Collapse
Affiliation(s)
- Aakanksha Jha
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742 USA
| | - Erika Moore
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742 USA
| |
Collapse
|
15
|
Lemaitre L, Adeniji N, Suresh A, Reguram R, Zhang J, Park J, Reddy A, Trevino AE, Mayer AT, Deutzmann A, Hansen AS, Tong L, Arjunan V, Kambham N, Visser BC, Dua MM, Bonham CA, Kothary N, D'Angio HB, Preska R, Rosen Y, Zou J, Charu V, Felsher DW, Dhanasekaran R. Spatial analysis reveals targetable macrophage-mediated mechanisms of immune evasion in hepatocellular carcinoma minimal residual disease. NATURE CANCER 2024; 5:1534-1556. [PMID: 39304772 DOI: 10.1038/s43018-024-00828-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 08/14/2024] [Indexed: 09/22/2024]
Abstract
Hepatocellular carcinoma (HCC) frequently recurs from minimal residual disease (MRD), which persists after therapy. Here, we identified mechanisms of persistence of residual tumor cells using post-chemoembolization human HCC (n = 108 patients, 1.07 million cells) and a transgenic mouse model of MRD. Through single-cell high-plex cytometric imaging, we identified a spatial neighborhood within which PD-L1 + M2-like macrophages interact with stem-like tumor cells, correlating with CD8+ T cell exhaustion and poor survival. Further, through spatial transcriptomics of residual HCC, we showed that macrophage-derived TGFβ1 mediates the persistence of stem-like tumor cells. Last, we demonstrate that combined blockade of Pdl1 and Tgfβ excluded immunosuppressive macrophages, recruited activated CD8+ T cells and eliminated residual stem-like tumor cells in two mouse models: a transgenic model of MRD and a syngeneic orthotopic model of doxorubicin-resistant HCC. Thus, our spatial analyses reveal that PD-L1+ macrophages sustain MRD by activating the TGFβ pathway in stem-like cancer cells and targeting this interaction may prevent HCC recurrence from MRD.
Collapse
Affiliation(s)
- Lea Lemaitre
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, USA
| | - Nia Adeniji
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, USA
| | - Akanksha Suresh
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, USA
| | - Reshma Reguram
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, USA
| | - Josephine Zhang
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, USA
| | - Jangho Park
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, USA
| | - Amit Reddy
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, USA
| | | | | | - Anja Deutzmann
- Division of Oncology, Departments of Medicine and Pathology, Stanford University, Stanford, CA, USA
| | - Aida S Hansen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Ling Tong
- Division of Oncology, Departments of Medicine and Pathology, Stanford University, Stanford, CA, USA
| | - Vinodhini Arjunan
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, USA
| | - Neeraja Kambham
- Department of Pathology, Stanford University, Stanford, CA, USA
| | | | - Monica M Dua
- Department of Surgery, Stanford University, Stanford, CA, USA
| | - C Andrew Bonham
- Department of Surgery, Stanford University, Stanford, CA, USA
| | - Nishita Kothary
- Department of Radiology, Stanford University, Stanford, CA, USA
| | | | | | - Yanay Rosen
- Department of Biomedical Data Science and Computer Science, Stanford University, Stanford, CA, USA
| | - James Zou
- Department of Biomedical Data Science and Computer Science, Stanford University, Stanford, CA, USA
| | - Vivek Charu
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Dean W Felsher
- Division of Oncology, Departments of Medicine and Pathology, Stanford University, Stanford, CA, USA.
| | | |
Collapse
|
16
|
Jacobs MME, Maas RJF, Jonkman I, Negishi Y, Tielemans Zamora W, Yanginlar C, van Heck J, Matzaraki V, Martens JHA, Baltissen M, Vermeulen M, Morla-Folch J, Ranzenigo A, Wang W, Umali M, Ochando J, van der Vlag J, Hilbrands LB, Joosten LAB, Netea MG, Mulder WJM, van Leent MMT, Mhlanga MM, Teunissen AJP, Rother N, Duivenvoorden R. Trained immunity is regulated by T cell-induced CD40-TRAF6 signaling. Cell Rep 2024; 43:114664. [PMID: 39178113 PMCID: PMC11536040 DOI: 10.1016/j.celrep.2024.114664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 07/08/2024] [Accepted: 08/06/2024] [Indexed: 08/25/2024] Open
Abstract
Trained immunity is characterized by histone modifications and metabolic changes in innate immune cells following exposure to inflammatory signals, leading to heightened responsiveness to secondary stimuli. Although our understanding of the molecular regulation of trained immunity has increased, the role of adaptive immune cells herein remains largely unknown. Here, we show that T cells modulate trained immunity via cluster of differentiation 40-tissue necrosis factor receptor-associated factor 6 (CD40-TRAF6) signaling. CD40-TRAF6 inhibition modulates functional, transcriptomic, and metabolic reprogramming and modifies histone 3 lysine 4 trimethylation associated with trained immunity. Besides in vitro studies, we reveal that single-nucleotide polymorphisms in the proximity of CD40 are linked to trained immunity responses in vivo and that combining CD40-TRAF6 inhibition with cytotoxic T lymphocyte antigen 4-immunoglobulin (CTLA4-Ig)-mediated co-stimulatory blockade induces long-term graft acceptance in a murine heart transplantation model. Combined, our results reveal that trained immunity is modulated by CD40-TRAF6 signaling between myeloid and adaptive immune cells and that this can be leveraged for therapeutic purposes.
Collapse
Affiliation(s)
- Maaike M E Jacobs
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Rianne J F Maas
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Inge Jonkman
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Yutaka Negishi
- Department of Cell Biology, Faculty of Science, Radboud University, Nijmegen, the Netherlands; Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Willem Tielemans Zamora
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cansu Yanginlar
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Julia van Heck
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Vasiliki Matzaraki
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Joost H A Martens
- Department of Molecular Biology, Faculty of Science, Oncode Institute, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Marijke Baltissen
- Department of Molecular Biology, Faculty of Science, Oncode Institute, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Michiel Vermeulen
- Department of Molecular Biology, Faculty of Science, Oncode Institute, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Judit Morla-Folch
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anna Ranzenigo
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - William Wang
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Martin Umali
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jordi Ochando
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Transplant Immunology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Johan van der Vlag
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Luuk B Hilbrands
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Medical Genetics, University of Medicine and Pharmacy, Iuliu Haţieganu, Cluj-Napoca, Romania
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Willem J M Mulder
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Mandy M T van Leent
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Musa M Mhlanga
- Department of Cell Biology, Faculty of Science, Radboud University, Nijmegen, the Netherlands; Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Abraham J P Teunissen
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nils Rother
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Raphaël Duivenvoorden
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands; BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
17
|
Marrufo AM, Flores-Mireles AL. Macrophage fate: to kill or not to kill? Infect Immun 2024; 92:e0047623. [PMID: 38829045 PMCID: PMC11385966 DOI: 10.1128/iai.00476-23] [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] [Indexed: 06/05/2024] Open
Abstract
Macrophages are dynamic innate immune cells that either reside in tissue, serving as sentinels, or recruited as monocytes from bone marrow into inflamed and infected tissue. In response to cues in the tissue microenvironment (TME), macrophages polarize on a continuum toward M1 or M2 with diverse roles in progression and resolution of disease. M1-like macrophages exhibit proinflammatory functions with antimicrobial and anti-tumorigenic activities, while M2-like macrophages have anti-inflammatory functions that generally resolve inflammatory responses and orchestrate a tissue healing process. Given these opposite phenotypes, proper spatiotemporal coordination of macrophage polarization in response to cues within the TME is critical to effectively resolve infectious disease and regulate wound healing. However, if this spatiotemporal coordination becomes disrupted due to persistent infection or dysregulated coagulation, macrophages' inappropriate response to these cues will result in the development of diseases with clinically unfavorable outcomes. Since plasticity and heterogeneity are hallmarks of macrophages, they are attractive targets for therapies to reprogram toward specific phenotypes that could resolve disease and favor clinical prognosis. In this review, we discuss how basic science studies have elucidated macrophage polarization mechanisms in TMEs during infections and inflammation, particularly coagulation. Therefore, understanding the dynamics of macrophage polarization within TMEs in diseases is important in further development of targeted therapies.
Collapse
Affiliation(s)
- Armando M. Marrufo
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | | |
Collapse
|
18
|
Fokra A, Feldman HB, Kurolap A, Kinaneh S, Abassi Z, Hershkovitz T. Patients with Gaucher disease display systemic elevation of ACE2, which is impacted by therapy status and genotype. Mol Genet Metab 2024; 143:108534. [PMID: 39033630 DOI: 10.1016/j.ymgme.2024.108534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/07/2024] [Accepted: 07/08/2024] [Indexed: 07/23/2024]
Abstract
Gaucher disease (GD) has a high carrier rate among Ashkenazi Jews.The most common disease-causing variant in this population N370S, is also prevalent pan-ethnically. This has led to speculations of some protective effect for carriers of this variant. During the recent COVID-19 pandemic, GD patients reportedly had a surprisingly low infection rate and mild symptoms considering their disease status. As SARS-CoV-2 gains entry into the cell via membrane-bound angiotensin-converting enzyme 2 (ACE2), we speculated that differences in levels of soluble ACE2 in GD patients could contribute to this protective state. While ACE is known to be elevated in GD, to our knowledge, ACE2 levels have not been explored. We measured serum and macrophage-bound levels of ACE and ACE2 by ELISA and western blot, respectively, in GD patients and age- and sex-matched controls. Our results reveal a significant elevation of both serum and macrophage-bound ACE and ACE2 in GD patients compared to healthy controls. This elevation appears to be mitigated by GD treatment. Moreover, the most robust ACE2 elevation was observed in N370S homozygotes, and was not effected by treatment. Since coronaviruses use the ACE2 receptor as a gateway for host cell entry, we speculate that elevated circulating ACE2 may serve as a decoy. This might explain the observed mild infections in GD patients during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Ahmad Fokra
- Department of Physiology and Biophysics Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Hagit Baris Feldman
- The Genetics Institute and Genomics Center, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Alina Kurolap
- The Genetics Institute and Genomics Center, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Safa Kinaneh
- Department of Physiology and Biophysics Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Zaid Abassi
- Department of Physiology and Biophysics Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel; Laboratory Medicine, Rambam Health Care Campus, Haifa, Israel
| | - Tova Hershkovitz
- Institute of Human Genetics, Galilee Medical Center, Nahariya, Israel.
| |
Collapse
|
19
|
Lewallen EA, Liu D, Karwoski J, Szeto WY, van Wijnen AJ, Laudanski K. Transcriptomic responses of peripheral blood leukocytes to cardiac surgery after acute inflammation, and three months recovery. Genomics 2024; 116:110878. [PMID: 38851465 DOI: 10.1016/j.ygeno.2024.110878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 04/29/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024]
Abstract
Traumatic perioperative conditions may trigger early systemic responses, activate leukocytes and reprogram the immune system. We hypothesize that leukocyte activation may not revert to pre-surgical states, and that protracted activation may emerge with increased risks of comorbidities. We tested this concept by examining the transcriptomes of monocytes and T cells in a representative observational cohort of patients (n = 13) admitted for elective cardiac surgery. Transcriptomes in T cells and monocytes were compared from before surgery (t0), and monocytes were analyzed longitudinally after acute (t24hr), and convalescent (t3m) time points. Monocytes and T cells expressed distinct transcriptomes, reflected by statistically significant differential expression of 558 T cell related genes. Monocytes expressed genes related to protein degradation and presented atypical activation of surface markers and cytoplasmic functions over time. Additionally, monocytes exhibited limited transcriptomic heterogeneity prior to surgery, and long-term patterns of gene expression associated with atherosclerosis showed three temporally distinct signatures. These data establish that post-cardiac surgery transcriptomes of monocytes differ even at three months compared to baselines, which may reflect latent ('smoldering') inflammation and persistent progression of tissue degenerative processes that should inform clinical care.
Collapse
Affiliation(s)
- Eric A Lewallen
- Department of Biological Sciences, Hampton University, Hampton, VA, USA.
| | - Da Liu
- Department of Obstetrics and Gynecology, Shengjin Hospital of China Medical University, Shenyang, Peoples Republic of China.
| | - Jake Karwoski
- Department of Undergraduate Studies, Drexel University, Philadelphia, PA, USA.
| | - Wilson Y Szeto
- Division of Cardiovascular Surgery, University of Pennsylvania, Philadelphia, PA, USA.
| | | | - Krzysztof Laudanski
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
20
|
Li J, Chen P, Ma W. The next frontier in immunotherapy: potential and challenges of CAR-macrophages. Exp Hematol Oncol 2024; 13:76. [PMID: 39103972 PMCID: PMC11302330 DOI: 10.1186/s40164-024-00549-9] [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: 06/04/2024] [Accepted: 07/30/2024] [Indexed: 08/07/2024] Open
Abstract
Chimeric antigen receptor macrophage (CAR-MΦ) represents a significant advancement in immunotherapy, especially for treating solid tumors where traditional CAR-T therapies face limitations. CAR-MΦ offers a promising approach to target and eradicate tumor cells by utilizing macrophages' phagocytic and antigen-presenting abilities. However, challenges such as the complex tumor microenvironment (TME), variability in antigen expression, and immune suppression limit their efficacy. This review addresses these issues, exploring mechanisms of CAR-MΦ action, optimal construct designs, and interactions within the TME. It also delves into the ex vivo manufacturing challenges of CAR-MΦ, discussing autologous and allogeneic sources and the importance of stringent quality control. The potential synergies of integrating CAR-MΦ with existing cancer therapies like checkpoint inhibitors and conventional chemotherapeutics are examined to highlight possible enhanced treatment outcomes. Furthermore, regulatory pathways for CAR-MΦ therapies are scrutinized alongside established protocols for CAR-T cells, identifying unique considerations essential for clinical trials and market approval. Proposed safety monitoring frameworks aim to manage potential adverse events, such as cytokine release syndrome, crucial for patient safety. Consolidating current research and clinical insights, this review seeks to refine CAR-MΦ therapeutic applications, overcome barriers, and suggest future research directions to transition CAR-MΦ therapies from experimental platforms to standard cancer care options.
Collapse
Affiliation(s)
- Jing Li
- The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, China
| | - Ping Chen
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Union Hospital, Fujian Medical University Fuzhou, Fujian, 350001, China
| | - Wenxue Ma
- Sanford Stem Cell Institute, Moores Cancer Center, University of California San Diego, CA, 92093, La Jolla, USA.
| |
Collapse
|
21
|
Schweiger P, Hamann L, Strobel J, Weisbach V, Wandersee A, Christ J, Kehl S, Weidenthaler F, Antoniadis S, Hackstein H, Cunningham S. Functional Heterogeneity of Umbilical Cord Blood Monocyte-Derived Dendritic Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:115-124. [PMID: 38809115 PMCID: PMC11215632 DOI: 10.4049/jimmunol.2400036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/14/2024] [Indexed: 05/30/2024]
Abstract
Human umbilical cord blood (UCB) represents a unique resource for hematopoietic stem cell transplantation for children and patients lacking suitable donors. UCB harbors a diverse set of leukocytes such as professional APCs, including monocytes, that could act as a novel source for cellular therapies. However, the immunological properties of UCB monocytes and monocyte-derived dendritic cells (MoDCs) are not fully characterized. In this study, we characterized the phenotype and functions of UCB-MoDCs to gauge their potential for future applications. UCB exhibited higher frequencies of platelets and lymphocytes as well as lower frequencies of neutrophils in comparison with adult whole blood. Leukocyte subset evaluation revealed significantly lower frequencies of granulocytes, NK cells, and CD14+CD16- monocytes. Surface marker evaluation revealed significantly lower rates of costimulatory molecules CD80 and CD83 while chemokine receptors CCR7 and CXCR4, as well as markers for Ag presentation, were similarly expressed. UCB-MoDCs were sensitive to TLR1-9 stimulation and presented quantitative differences in the release of proinflammatory cytokines. UCB-MoDCs presented functional CCR7-, CXCR4-, and CCR5-associated migratory behavior as well as adequate receptor- and micropinocytosis-mediated Ag uptake. When cocultured with allogeneic T lymphocytes, UCB-MoDCs induced weak CD4+ T lymphocyte proliferation, CD71 expression, and release of IFN-γ and IL-2. Taken together, UCB-MoDCs present potentially advantageous properties for future medical applications.
Collapse
Affiliation(s)
- Petra Schweiger
- Department of Transfusion Medicine and Haemostaseology, Friedrich–Alexander University of Erlangen–Nuremberg, University Hospital of Erlangen, Erlangen, Germany
| | - Livia Hamann
- Department of Transfusion Medicine and Haemostaseology, Friedrich–Alexander University of Erlangen–Nuremberg, University Hospital of Erlangen, Erlangen, Germany
| | - Julian Strobel
- Department of Transfusion Medicine and Haemostaseology, Friedrich–Alexander University of Erlangen–Nuremberg, University Hospital of Erlangen, Erlangen, Germany
| | - Volker Weisbach
- Department of Transfusion Medicine and Haemostaseology, Friedrich–Alexander University of Erlangen–Nuremberg, University Hospital of Erlangen, Erlangen, Germany
| | - Alexandra Wandersee
- Department of Transfusion Medicine and Haemostaseology, Friedrich–Alexander University of Erlangen–Nuremberg, University Hospital of Erlangen, Erlangen, Germany
| | - Julia Christ
- Department of Transfusion Medicine and Haemostaseology, Friedrich–Alexander University of Erlangen–Nuremberg, University Hospital of Erlangen, Erlangen, Germany
| | - Sven Kehl
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Erlangen, Germany
| | - Filip Weidenthaler
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Erlangen, Germany
| | - Sophia Antoniadis
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Erlangen, Germany
| | - Holger Hackstein
- Department of Transfusion Medicine and Haemostaseology, Friedrich–Alexander University of Erlangen–Nuremberg, University Hospital of Erlangen, Erlangen, Germany
| | - Sarah Cunningham
- Department of Transfusion Medicine and Haemostaseology, Friedrich–Alexander University of Erlangen–Nuremberg, University Hospital of Erlangen, Erlangen, Germany
| |
Collapse
|
22
|
Campo M, Dill-McFarland KA, Peterson GJ, Benson B, Skerrett SJ, Hawn TR. Human Alveolar and Monocyte-Derived Human Macrophage Responses to Mycobacterium tuberculosis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:161-169. [PMID: 38836816 PMCID: PMC11610518 DOI: 10.4049/jimmunol.2300885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 04/29/2024] [Indexed: 06/06/2024]
Abstract
Alveolar macrophages (AMs) and recruited monocyte-derived macrophages (MDMs) mediate early lung immune responses to Mycobacterium tuberculosis. Differences in the response of these distinct cell types are poorly understood and may provide insight into mechanisms of tuberculosis pathogenesis. The objective of this study was to determine whether M. tuberculosis induces unique and essential antimicrobial pathways in human AMs compared with MDMs. Using paired human AMs and 5-d MCSF-derived MDMs from six healthy volunteers, we infected cells with M. tuberculosis H37Rv for 6 h, isolated RNA, and analyzed transcriptomic profiles with RNA sequencing. We found 681 genes that were M. tuberculosis dependent in AMs compared with MDMs and 4538 that were M. tuberculosis dependent in MDMs, but not AMs (false discovery rate [FDR] < 0.05). Using hypergeometric enrichment of DEGs in Broad Hallmark gene sets, we found that type I and II IFN Response were the only gene sets selectively induced in M. tuberculosis-infected AM (FDR < 0.05). In contrast, MYC targets, unfolded protein response and MTORC1 signaling, were selectively enriched in MDMs (FDR < 0.05). IFNA1, IFNA8, IFNE, and IFNL1 were specifically and highly upregulated in AMs compared with MDMs at baseline and/or after M. tuberculosis infection. IFNA8 modulated M. tuberculosis-induced proinflammatory cytokines and, compared with other IFNs, stimulated unique transcriptomes. Several DNA sensors and IFN regulatory factors had higher expression at baseline and/or after M. tuberculosis infection in AMs compared with MDMs. These findings demonstrate that M. tuberculosis infection induced unique transcriptional responses in human AMs compared with MDMs, including upregulation of the IFN response pathway and specific DNA sensors.
Collapse
Affiliation(s)
- Monica Campo
- Department of Medicine, University of Minnesota, Minneapolis, MN
| | | | | | - Basilin Benson
- Systems Immunology Program, Benaroya Research Institute, Seattle, WA
| | | | - Thomas R. Hawn
- Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|
23
|
Chini A, Guha P, Rishi A, Obaid M, Udden SN, Mandal SS. Discovery and functional characterization of LncRNAs associated with inflammation and macrophage activation. Methods 2024; 227:1-16. [PMID: 38703879 DOI: 10.1016/j.ymeth.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/24/2024] [Accepted: 05/01/2024] [Indexed: 05/06/2024] Open
Abstract
Long noncoding RNAs (lncRNA) are emerging players in regulation of gene expression and cell signaling and their dysregulation has been implicated in a multitude of human diseases. Recent studies from our laboratory revealed that lncRNAs play critical roles in cytokine regulation, inflammation, and metabolism. We demonstrated that lncRNA HOTAIR, which is a well-known regulator of gene silencing, plays critical roles in modulation of cytokines and proinflammatory genes, and glucose metabolism in macrophages during inflammation. In addition, we recently discovered a series of novel lncRNAs that are closely associated with inflammation and macrophage activation. We termed these as long-noncoding inflammation associated RNAs (LinfRNAs). We are currently engaged in the functional characterization of these hLinfRNAs (human LinfRNAs) with a focus on their roles in inflammation, and we are investigating their potential implications in chronic inflammatory human diseases. Here, we have summarized experimental methods that have been utilized for the discovery and functional characterization of lncRNAs in inflammation and macrophage activation.
Collapse
Affiliation(s)
- Avisankar Chini
- Gene Regulation and Epigenetics Research Laboratory, Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Prarthana Guha
- Gene Regulation and Epigenetics Research Laboratory, Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Ashcharya Rishi
- Gene Regulation and Epigenetics Research Laboratory, Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Monira Obaid
- Gene Regulation and Epigenetics Research Laboratory, Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Sm Nashir Udden
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Subhrangsu S Mandal
- Gene Regulation and Epigenetics Research Laboratory, Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX 76019, USA.
| |
Collapse
|
24
|
David CAW, Vermeulen JP, Gioria S, Vandebriel RJ, Liptrott NJ. Nano(bio)Materials Do Not Affect Macrophage Phenotype-A Study Conducted by the REFINE Project. Int J Mol Sci 2024; 25:5491. [PMID: 38791527 PMCID: PMC11121830 DOI: 10.3390/ijms25105491] [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: 04/03/2024] [Revised: 05/10/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024] Open
Abstract
Macrophages are well known for their involvement in the biocompatibility, as well as biodistribution, of nano(bio)materials. Although there are a number of rodent cell lines, they may not fully recapitulate primary cell responses, particularly those of human cells. Isolation of tissue-resident macrophages from humans is difficult and may result in insufficient cells with which to determine the possible interaction with nano(bio)materials. Isolation of primary human monocytes and differentiation to monocyte-derived macrophages may provide a useful tool with which to further study these interactions. To that end, we developed a standard operating procedure for this differentiation, as part of the Regulatory Science Framework for Nano(bio)material-based Medical Products and Devices (REFINE) project, and used it to measure the secretion of bioactive molecules from M1 and M2 differentiated monocytes in response to model nano(bio)materials, following an initial assessment of pyrogenic contamination, which may confound potential observations. The SOP was deployed in two partner institutions with broadly similar results. The work presented here shows the utility of this assay but highlights the relevance of donor variability in responses to nano(bio)materials. Whilst donor variability can provide some logistical challenges to the application of such assays, this variability is much closer to the heterogeneous cells that are present in vivo, compared to homogeneous non-human cell lines.
Collapse
Affiliation(s)
- Christopher A. W. David
- Immunocompatibility Group, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L7 3NY, UK;
- Centre of Excellence for Long-Acting Therapeutics (CELT), Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L7 8TX, UK
| | - Jolanda P. Vermeulen
- National Institute for Public Health & the Environment, 3720 BA Bilthoven, The Netherlands; (J.P.V.); (R.J.V.)
| | - Sabrina Gioria
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy;
| | - Rob J. Vandebriel
- National Institute for Public Health & the Environment, 3720 BA Bilthoven, The Netherlands; (J.P.V.); (R.J.V.)
| | - Neill J. Liptrott
- Immunocompatibility Group, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L7 3NY, UK;
- Centre of Excellence for Long-Acting Therapeutics (CELT), Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L7 8TX, UK
| |
Collapse
|
25
|
Gilchrist JJ, Fang H, Danielli S, Tomkova M, Nassiri I, Ng E, Tong O, Taylor C, Muldoon D, Cohen LRZ, Al-Mossawi H, Lau E, Neville M, Schuster-Boeckler B, Knight JC, Fairfax BP. Characterization of the genetic determinants of context-specific DNA methylation in primary monocytes. CELL GENOMICS 2024; 4:100541. [PMID: 38663408 PMCID: PMC11099345 DOI: 10.1016/j.xgen.2024.100541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 11/24/2023] [Accepted: 03/27/2024] [Indexed: 05/12/2024]
Abstract
To better understand inter-individual variation in sensitivity of DNA methylation (DNAm) to immune activity, we characterized effects of inflammatory stimuli on primary monocyte DNAm (n = 190). We find that monocyte DNAm is site-dependently sensitive to lipopolysaccharide (LPS), with LPS-induced demethylation occurring following hydroxymethylation. We identify 7,359 high-confidence immune-modulated CpGs (imCpGs) that differ in genomic localization and transcription factor usage according to whether they represent a gain or loss in DNAm. Demethylated imCpGs are profoundly enriched for enhancers and colocalize to genes enriched for disease associations, especially cancer. DNAm is age associated, and we find that 24-h LPS exposure triggers approximately 6 months of gain in epigenetic age, directly linking epigenetic aging with innate immune activity. By integrating LPS-induced changes in DNAm with genetic variation, we identify 234 imCpGs under local genetic control. Exploring shared causal loci between LPS-induced DNAm responses and human disease traits highlights examples of disease-associated loci that modulate imCpG formation.
Collapse
Affiliation(s)
- James J Gilchrist
- Department of Paediatrics, University of Oxford, Oxford OX3 9DU, UK; MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Hai Fang
- Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Sara Danielli
- Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Marketa Tomkova
- Ludwig Cancer Research Oxford, University of Oxford, Oxford OX3 7DQ, UK
| | - Isar Nassiri
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK; Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Esther Ng
- Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK
| | - Orion Tong
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Chelsea Taylor
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Dylan Muldoon
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Lea R Z Cohen
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Hussein Al-Mossawi
- Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK
| | - Evelyn Lau
- Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Matt Neville
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LE, UK
| | | | - Julian C Knight
- Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Benjamin P Fairfax
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK; Department of Oncology, University of Oxford, Oxford OX3 9DS, UK.
| |
Collapse
|
26
|
Szigeti K, Ihnatovych I, Notari E, Dorn RP, Maly I, He M, Birkaya B, Prasad S, Byrne RS, Indurthi DC, Nimmer E, Heo Y, Retfalvi K, Chaves L, Sule N, Hofmann WA, Auerbach A, Wilding G, Bae Y, Reynolds J. CHRFAM7A diversifies human immune adaption through Ca 2+ signalling and actin cytoskeleton reorganization. EBioMedicine 2024; 103:105093. [PMID: 38569318 PMCID: PMC10999709 DOI: 10.1016/j.ebiom.2024.105093] [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: 10/10/2023] [Revised: 03/15/2024] [Accepted: 03/17/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Human restricted genes contribute to human specific traits in the immune system. CHRFAM7A, a uniquely human fusion gene, is a negative regulator of the α7 nicotinic acetylcholine receptor (α7 nAChR), the highest Ca2+ conductor of the ACh receptors implicated in innate immunity. Understanding the mechanism of how CHRFAM7A affects the immune system remains unexplored. METHODS Two model systems are used, human induced pluripotent stem cells (iPSC) and human primary monocytes, to characterize α7 nAChR function, Ca2+ dynamics and decoders to elucidate the pathway from receptor to phenotype. FINDINGS CHRFAM7A/α7 nAChR is identified as a hypomorphic receptor with mitigated Ca2+ influx and prolonged channel closed state. This shifts the Ca2+ reservoir from the extracellular space to the endoplasmic reticulum (ER) leading to Ca2+ dynamic changes. Ca2+ decoder small GTPase Rac1 is then activated, reorganizing the actin cytoskeleton. Observed actin mediated phenotypes include cellular adhesion, motility, phagocytosis and tissue mechanosensation. INTERPRETATION CHRFAM7A introduces an additional, human specific, layer to Ca2+ regulation leading to an innate immune gain of function. Through the actin cytoskeleton it drives adaptation to the mechanical properties of the tissue environment leading to an ability to invade previously immune restricted niches. Human genetic diversity predicts profound translational significance as its understanding builds the foundation for successful treatments for infectious diseases, sepsis, and cancer metastasis. FUNDING This work is supported in part by the Community Foundation for Greater Buffalo (Kinga Szigeti) and in part by NIH grant R01HL163168 (Yongho Bae).
Collapse
Affiliation(s)
- Kinga Szigeti
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA.
| | - Ivanna Ihnatovych
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Emily Notari
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Ryu P Dorn
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Ivan Maly
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Muye He
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Barbara Birkaya
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Shreyas Prasad
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Robin Schwartz Byrne
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Dinesh C Indurthi
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Erik Nimmer
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Yuna Heo
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Kolos Retfalvi
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Lee Chaves
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Norbert Sule
- Roswell Park Comprehensive Cancer Center, 665 Elm St, Buffalo, NY, 14203, USA
| | - Wilma A Hofmann
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Anthony Auerbach
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Gregory Wilding
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Yongho Bae
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Jessica Reynolds
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| |
Collapse
|
27
|
Jesus S, Panão Costa J, Colaço M, Lebre F, Mateus D, Sebastião AI, Cruz MT, Alfaro-Moreno E, Borges O. Exploring the immunomodulatory properties of glucan particles in human primary cells. Int J Pharm 2024; 655:123996. [PMID: 38490404 DOI: 10.1016/j.ijpharm.2024.123996] [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: 11/15/2023] [Revised: 03/05/2024] [Accepted: 03/11/2024] [Indexed: 03/17/2024]
Abstract
The immunomodulatory properties of β-glucans have sparked interest among various medical fields. As vaccine adjuvants, glucan particles offer additional advantages as antigen delivery systems. This study reported the immunomodulatory properties of glucan particles with different size and chemical composition. The effect of glucan microparticles (GPs) and glucan nanoparticles (Glu 130 and 355 NPs) was evaluated on human immune cells. While GPs and Glu 355 NPs demonstrated substantial interaction with Dectin-1 receptor on monocytes, Glu 130 NPs exhibited reduced activation of this receptor. This observation was substantiated by blocking Dectin-1, resulting in inhibition of reactive oxygen species production induced by GPs and Glu 355 NPs. Notably, monocyte-derived dendritic cells (moDCs) stimulated by Glu 355 NPs exhibited phenotypic and functional maturation, essential for antigen cross-presentation. The immunomodulatory efficacy was investigated using an autologous mixed lymphocyte reaction (AMLR), resulting in considerable rates of lymphocyte proliferation and an intriguing profile of cytokine and chemokine release. Our findings highlight the importance of meticulously characterizing the size and chemical composition of β-glucan particles to draw accurate conclusions regarding their immunomodulatory activity. This in vitro model mimics the human cellular immune response, and the results obtained endorse the use of β-glucan-based delivery systems as future vaccine adjuvants.
Collapse
Affiliation(s)
- Sandra Jesus
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal
| | - João Panão Costa
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Portugal
| | - Mariana Colaço
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Portugal
| | - Filipa Lebre
- Nanosafety Group, International Iberian Nanotechnology Laboratory, Braga, Portugal
| | - Daniela Mateus
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Portugal
| | - Ana Isabel Sebastião
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Portugal
| | - Maria T Cruz
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Portugal
| | | | - Olga Borges
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Portugal.
| |
Collapse
|
28
|
Xu JC, Wu K, Ma RQ, Li JH, Tao J, Hu Z, Fan XY. Establishment of an in vitro model of monocyte-like THP-1 cells for trained immunity induced by bacillus Calmette-Guérin. BMC Microbiol 2024; 24:130. [PMID: 38643095 PMCID: PMC11031977 DOI: 10.1186/s12866-024-03191-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: 06/19/2023] [Accepted: 01/10/2024] [Indexed: 04/22/2024] Open
Abstract
BACKGROUND Mycobacteria bloodstream infections are common in immunocompromised people and usually have disastrous consequences. As the primary phagocytes in the bloodstream, monocytes and neutrophils play critical roles in the fight against bloodstream mycobacteria infections. In contrast to macrophages, the responses of monocytes infected with the mycobacteria have been less investigated. RESULTS In this study, we first established a protocol for infection of non-adherent monocyte-like THP-1 cells (i.e. without the differentiation induced by phorbol 12-myristate 13-acetate (PMA) by bacillus Calmette-Guérin (BCG). Via the protocol, we were then capable of exploring the global transcriptomic profiles of non-adherent THP-1 cells infected with BCG, and found that NF-κB, MAPK and PI3K-Akt signaling pathways were enhanced, as well as some inflammatory chemokine/cytokine genes (e.g. CCL4, CXCL10, TNF and IL-1β) were up-regulated. Surprisingly, the Akt-HIF-mTOR signaling pathway was also activated, which induces trained immunity. In this in vitro infection model, increased cytokine responses to lipopolysaccharides (LPS) restimulation, higher cell viability, and decreased Candida albicans loads were observed. CONCLUSIONS We have first characterized the transcriptomic profiles of BCG-infected non-adherent THP-1 cells, and first developed a trained immunity in vitro model of the cells.
Collapse
Affiliation(s)
- Jin-Chuan Xu
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai, China
| | - Kang Wu
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai, China
- Shanghai R & S Biotech. Co., Ltd, Shanghai, China
- Zhejiang Free Trade Area R & S Biomedical Technology Co., Ltd, Zhoushan, Zhejiang, China
| | - Rui-Qing Ma
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai, China
| | - Jian-Hui Li
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai, China
| | - Jie Tao
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai, China
| | - Zhidong Hu
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai, China
| | - Xiao-Yong Fan
- Shanghai Public Health Clinical Center & Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai, China.
| |
Collapse
|
29
|
Padovani CM, Wilson RM, Rodriguez A, Spur BW, Yin K. Resolvin D2 attenuates LPS-induced macrophage exhaustion. FASEB J 2024; 38:e23569. [PMID: 38551610 DOI: 10.1096/fj.202302521r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/21/2024] [Accepted: 03/07/2024] [Indexed: 04/02/2024]
Abstract
Early in sepsis, a hyperinflammatory response is dominant, but later, an immunosuppressive phase dominates, and the host is susceptible to opportunistic infections. Anti-inflammatory agents may accelerate the host into immunosuppression, and few agents can reverse immunosuppression without causing inflammation. Specialized pro-resolving mediators (SPMs) such as resolvin D2 (RvD2) have been reported to resolve inflammation without being immunosuppressive, but little work has been conducted to examine their effects on immunosuppression. To assess the effects of RvD2 on immunosuppression, we established a model of macrophage exhaustion using two lipopolysaccharide (LPS) treatments or hits. THP-1 monocyte-derived macrophages were first treated with RvD2 or vehicle for 1 h. One LPS hit increased NF-κB activity 11-fold and TNF-α release 60-fold compared to unstimulated macrophages. RvD2 decreased LPS-induced NF-κB activity and TNF-α production but increased bacterial clearance. Two LPS hits reduced macrophage bacterial clearance and decreased macrophage NF-κB activity (45%) and TNF-α release (75%) compared to one LPS hit, demonstrating exhaustion. RvD2 increased NF-κB activity, TNF-α release, and bacterial clearance following two LPS hits compared to controls. TLR2 inhibition abolished RvD2-mediated changes. In a mouse sepsis model, splenic macrophage response to exogenous LPS was reduced compared to controls and was restored by in vivo administration of RvD2, supporting the in vitro results. If RvD2 was added to monocytes before differentiation into macrophages, however, RvD2 reduced LPS responses and increased bacterial clearance following both one and two LPS hits. The results show that RvD2 attenuated macrophage suppression in vitro and in vivo and that this effect was macrophage-specific.
Collapse
Affiliation(s)
- Cristina M Padovani
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Life Sciences of Rowan University, Stratford, New Jersey, USA
| | - Rachael M Wilson
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Life Sciences of Rowan University, Stratford, New Jersey, USA
| | - Ana Rodriguez
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Life Sciences of Rowan University, Stratford, New Jersey, USA
| | - Bernd W Spur
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Life Sciences of Rowan University, Stratford, New Jersey, USA
| | - Kingsley Yin
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Life Sciences of Rowan University, Stratford, New Jersey, USA
| |
Collapse
|
30
|
Okada T, Okada A, Aoki H, Onozato D, Kato T, Takase H, Ohshima S, Sugino T, Unno R, Taguchi K, Hamamoto S, Ando R, Shimada IS, Hashita T, Iwao T, Matsunaga T, Yasui T. Phagocytosis model of calcium oxalate monohydrate crystals generated using human induced pluripotent stem cell-derived macrophages. Urolithiasis 2024; 52:51. [PMID: 38554162 DOI: 10.1007/s00240-024-01553-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/03/2024] [Indexed: 04/01/2024]
Abstract
Macrophages play a role in nephrolithiasis, offering the possibility of developing macrophage-mediated preventive therapies. To establish a system for screening drugs that could prevent the formation of kidney stones, we aimed to develop a model using human induced pluripotent stem cell (iPSC)-derived macrophages to study phagocytosis of calcium oxalate monohydrate (COM) crystals. Human iPSCs (201B7) were cultured. CD14+ monocytes were recovered using a stepwise process that involved the use of growth factors and cytokines. These cells were then allowed to differentiate into M1 and M2 macrophages. The macrophages were co-cultured with COM crystals and used in the phagocytosis experiments. Live cell imaging and polarized light observation via super-resolution microscopy were used to visualize phagocytosis. Localization of phagocytosed COM crystals was observed using transmission electron microscopy. Intracellular fluorescence intensity was measured using imaging cytometry to quantify phagocytosis. Human iPSCs successfully differentiated into M1 and M2 macrophages. M1 macrophages adhered to the culture plate and moved COM crystals from the periphery to cell center over time, whereas M2 macrophages did not adhere to the culture plate and actively phagocytosed the surrounding COM crystals. Fluorescence assessment over a 24-h period showed that M2 macrophages exhibited higher intracellular fluorescence intensity (5.65-times higher than that of M1 macrophages at 4.5 h) and maintained this advantage for 18 h. This study revealed that human iPSC-derived macrophages have the ability to phagocytose COM crystals, presenting a new approach for studying urinary stone formation and highlighting the potential of iPSC-derived macrophages as a tool to screen nephrolithiasis-related drugs.
Collapse
Affiliation(s)
- Tomoki Okada
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| | - Atsushi Okada
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan.
| | - Hiromasa Aoki
- Department of Pathobiology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, Japan
| | - Daichi Onozato
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, Japan
| | - Taiki Kato
- Department of Urology, Nagoya City East Medical Center, Nagoya, Aichi, Japan
| | - Hiroshi Takase
- Core Laboratory, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Shigeru Ohshima
- Department of Medical Technology, Yokkaichi Nursing and Medical Technology school of Nursing and Medical Care, Yokkaichi, Mie, Japan
| | - Teruaki Sugino
- Department of Urology, Nagoya City East Medical Center, Nagoya, Aichi, Japan
| | - Rei Unno
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| | - Kazumi Taguchi
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| | - Shuzo Hamamoto
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| | - Ryosuke Ando
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| | - Issei S Shimada
- Department of Cell Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Tadahiro Hashita
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, Japan
| | - Takahiro Iwao
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, Japan
| | - Tamihide Matsunaga
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, Japan
| | - Takahiro Yasui
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| |
Collapse
|
31
|
Fageräng B, Lau C, Mc Adam KE, Schjalm C, Christiansen D, Garred P, Nilsson PH, Mollnes TE. A novel selective leukocyte depletion human whole blood model reveals the specific roles of monocytes and granulocytes in the cytokine response to Escherichia coli. J Leukoc Biol 2024; 115:647-663. [PMID: 38057165 DOI: 10.1093/jleuko/qiad151] [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: 05/19/2023] [Revised: 10/28/2023] [Accepted: 11/19/2023] [Indexed: 12/08/2023] Open
Abstract
The lepirudin-based human whole blood model is a well-established ex vivo system to characterize inflammatory responses. However, the contribution of individual cell populations to cytokine release has not been investigated. Thus, we modified the model by selectively removing leukocyte subpopulations to elucidate their contribution to the inflammatory response. Lepirudin-anticoagulated whole blood was depleted from monocytes or granulocytes using StraightFrom Whole Blood MicroBeads. Reconstituted blood was incubated with Escherichia coli (108/mL) for 2 hours at 37 °C. CD11b, CD62P, and CD63 were detected by flow cytometry. Complement (C3bc, sC5b-9) and platelet activation (platelet factor 4, NAP-2) were measured by enzyme-linked immunosorbent assay. Cytokines were quantified by multiplex assay. A significant (P < 0.05) specific depletion of the monocyte (mean = 86%; 95% confidence interval = 71%-92%) and granulocyte (mean = 97%; 95% confidence interval = 96%-98%) population was obtained. Background activation induced by the depletion protocol was negligible for complement (C3bc and sC5b-9), leukocytes (CD11b), and platelets (NAP-2). Upon Escherichia coli incubation, release of 10 of the 24 cytokines was solely dependent on monocytes (interleukin [IL]-1β, IL-2, IL-4, IL-5, IL-17A, interferon-γ, granulocyte colony-stimulating factor, granulocyte-macrophage colony-stimulating factor, macrophage inflammatory protein-1α, and fibroblast growth factor-basic), whereas 8 were dependent on both monocytes and granulocytes (IL-1ra, IL-6, IL-8, IL-9, IL-10, macrophage inflammatory protein-1β, tumor necrosis factor, and eotaxin). Six cytokines were not monocyte or granulocyte dependent, of which platelet-derived growth factor and RANTES were mainly platelet dependent. We document an effective model for selective depletion of leukocyte subpopulations from whole blood, without causing background activation, allowing in-depth cellular characterization. The results are in accordance with monocytes playing a major role in cytokine release and expand our knowledge of the significant role of granulocytes in the response to E. coli.
Collapse
Affiliation(s)
- Beatrice Fageräng
- Department of Immunology, Oslo University Hospital, University of Oslo, Sognsvannsveien 20, 0372 Oslo, Norway
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Ole Maaloesvej 26, 2200 Copenhagen, Denmark
| | - Corinna Lau
- Research Laboratory, Nordland Hospital, Parkveien 95, 8005 Bodø, Norway
| | - Karin Ekholt Mc Adam
- Department of Immunology, Oslo University Hospital, University of Oslo, Sognsvannsveien 20, 0372 Oslo, Norway
| | - Camilla Schjalm
- Department of Immunology, Oslo University Hospital, University of Oslo, Sognsvannsveien 20, 0372 Oslo, Norway
| | | | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Ole Maaloesvej 26, 2200 Copenhagen, Denmark
| | - Per H Nilsson
- Department of Immunology, Oslo University Hospital, University of Oslo, Sognsvannsveien 20, 0372 Oslo, Norway
- Linnæus Center of Biomaterials Chemistry, Linnæus University, Universitetsplatsen 1, 391 82 Kalmar, Sweden
- Department of Chemistry and Biomedical Sciences, Linnaeus University, Universitetsplatsen 1, 391 82 Kalmar, Sweden
| | - Tom Eirik Mollnes
- Department of Immunology, Oslo University Hospital, University of Oslo, Sognsvannsveien 20, 0372 Oslo, Norway
- Research Laboratory, Nordland Hospital, Parkveien 95, 8005 Bodø, Norway
| |
Collapse
|
32
|
Chandrabalan S, Dang L, Hansen U, Timmen M, Wehmeyer C, Stange R, Beißbarth T, Binder C, Bleckmann A, Menck K. A novel method to efficiently differentiate human osteoclasts from blood-derived monocytes. Biol Proced Online 2024; 26:7. [PMID: 38504200 PMCID: PMC10949786 DOI: 10.1186/s12575-024-00233-6] [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: 08/31/2023] [Accepted: 03/09/2024] [Indexed: 03/21/2024] Open
Abstract
BACKGROUND Osteoclasts are the tissue-specific macrophage population of the bone and unique in their bone-resorbing activity. Hence, they are fundamental for bone physiology in health and disease. However, efficient protocols for the isolation and study of primary human osteoclasts are scarce. In this study, we aimed to establish a protocol, which enables the efficient differentiation of functional human osteoclasts from monocytes. RESULTS Human monocytes were isolated through a double-density gradient from donor blood. Compared to standard differentiation schemes in polystyrene cell culture dishes, the yield of multinuclear osteoclasts was significantly increased upon initial differentiation of monocytes to macrophages in fluorinated ethylene propylene (FEP) Teflon bags. This initial differentiation phase was then followed by the development of terminal osteoclasts by addition of Receptor Activator of NF-κB Ligand (RANKL). High concentrations of RANKL and Macrophage colony-stimulating factor (M-CSF) as well as an intermediate cell density further supported efficient cell differentiation. The generated cells were highly positive for CD45, CD14 as well as the osteoclast markers CD51/ITGAV and Cathepsin K/CTSK, thus identifying them as osteoclasts. The bone resorption of the osteoclasts was significantly increased when the cells were differentiated from macrophages derived from Teflon bags compared to macrophages derived from conventional cell culture plates. CONCLUSION Our study has established a novel protocol for the isolation of primary human osteoclasts that improves osteoclastogenesis in comparison to the conventionally used cultivation approach.
Collapse
Affiliation(s)
- Suganja Chandrabalan
- Department of Medicine A, Hematology, Oncology, and Pneumology, University of Muenster, Muenster, Germany
- West German Cancer Center, University Hospital Muenster, Muenster, Germany
| | - Linh Dang
- Department of Medical Bioinformatics, University Medical Center Goettingen, Goettingen, Germany
| | - Uwe Hansen
- Institute of Musculoskeletal Medicine (IMM), University of Muenster, Muenster, Germany
| | - Melanie Timmen
- Institute of Musculoskeletal Medicine (IMM), University of Muenster, Muenster, Germany
| | - Corinna Wehmeyer
- Institute of Musculoskeletal Medicine (IMM), University of Muenster, Muenster, Germany
| | - Richard Stange
- Institute of Musculoskeletal Medicine (IMM), University of Muenster, Muenster, Germany
| | - Tim Beißbarth
- Department of Medical Bioinformatics, University Medical Center Goettingen, Goettingen, Germany
| | - Claudia Binder
- Department of Hematology/Medical Oncology, University Medical Center Goettingen, Goettingen, Germany
| | - Annalen Bleckmann
- Department of Medicine A, Hematology, Oncology, and Pneumology, University of Muenster, Muenster, Germany
- West German Cancer Center, University Hospital Muenster, Muenster, Germany
| | - Kerstin Menck
- Department of Medicine A, Hematology, Oncology, and Pneumology, University of Muenster, Muenster, Germany.
- West German Cancer Center, University Hospital Muenster, Muenster, Germany.
| |
Collapse
|
33
|
Szymczak B, Junkuszew A, Patkowski K, Szponder T, Ngoc DN, Drzewiecka B, Sobczyńska-Rak A, Wessely-Szponder J. The activity of monocyte-derived macrophages after stimulation with platelet-rich and platelet-poor concentrates. Study on an ovine model of insertion of a tibial implant coated with silicon-doped diamond-like carbon. J Vet Res 2024; 68:167-174. [PMID: 38525222 PMCID: PMC10960256 DOI: 10.2478/jvetres-2024-0003] [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: 07/28/2023] [Accepted: 01/15/2024] [Indexed: 03/26/2024] Open
Abstract
Introduction Macrophages are crucial immune cells that play a role in tissue repair and can exhibit pro- or anti-inflammatory behaviour based on environmental stimulation. Their functional phenotype can be affected by platelet-derived products as determined by those products' composition. When the inflammatory response caused by implantation is excessive, it can lead to rejection of the implant. Therefore, a thorough evaluation of implant haemocompatibility is necessary to minimise undesirable consequences. Material and Methods In an in vitro study, monocyte-derived macrophages (MDMs) were obtained from the whole blood of sheep after a silicon-doped diamond-like carbon-coated implant insertion. These MDMs were then exposed to autologous platelet-derived products for functional marker analysis. Results Platelet-poor plasma (PPP) and pure platelet-rich plasma (P-PRP) stimulation increased arginase-1 activity, while leukocyte-rich PRP stimulation produced a mixed response involving higher O2- (6.49 ± 2.43 nM vs non-stimulated 3.51 ± 1.23 nM, P-value < 0.05) and NO (3.28 ± 1.38 μM vs non-stimulated 2.55 ± 0.32μM, P-value < 0.05) generation. Conclusion Using PPP and P-PRP stimulation in post-implantation procedures may contribute to the polarisation of macrophages towards the M2-like pro-resolving phenotype, thereby accelerating wound healing. This would also prevent implant degradation due to an excessive inflammatory process.
Collapse
Affiliation(s)
- Bartłomiej Szymczak
- Sub-Department of Pathophysiology, Department of Preclinical Veterinary Sciences, University of Life Sciences, 20-950Lublin, Poland
| | - Andrzej Junkuszew
- Department of Animal Breeding and Agricultural Consulting, Faculty of Animal Sciences and Bioeconomy, University of Life Sciences, 20-950Lublin, Poland
| | - Krzysztof Patkowski
- Department of Animal Breeding and Agricultural Consulting, Faculty of Animal Sciences and Bioeconomy, University of Life Sciences, 20-950Lublin, Poland
| | - Tomasz Szponder
- Department and Clinic of Animal Surgery, Faculty of Veterinary Medicine, University of Life Sciences, 20-950Lublin, Poland
| | - Dominika Nguyen Ngoc
- Sub-Department of Pathophysiology, Department of Preclinical Veterinary Sciences, University of Life Sciences, 20-950Lublin, Poland
| | - Beata Drzewiecka
- Sub-Department of Pathophysiology, Department of Preclinical Veterinary Sciences, University of Life Sciences, 20-950Lublin, Poland
| | - Aleksandra Sobczyńska-Rak
- Department and Clinic of Animal Surgery, Faculty of Veterinary Medicine, University of Life Sciences, 20-950Lublin, Poland
| | - Joanna Wessely-Szponder
- Sub-Department of Pathophysiology, Department of Preclinical Veterinary Sciences, University of Life Sciences, 20-950Lublin, Poland
| |
Collapse
|
34
|
Yang S, Wang M, Hua Y, Li J, Zheng H, Cui M, Huang N, Liu Q, Liao Q. Advanced insights on tumor-associated macrophages revealed by single-cell RNA sequencing: The intratumor heterogeneity, functional phenotypes, and cellular interactions. Cancer Lett 2024; 584:216610. [PMID: 38244910 DOI: 10.1016/j.canlet.2024.216610] [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: 11/23/2022] [Revised: 11/28/2023] [Accepted: 12/18/2023] [Indexed: 01/22/2024]
Abstract
Single-cell RNA sequencing (scRNA-seq) is an emerging technology used for cellular transcriptome analysis. The application of scRNA-seq has led to profoundly advanced oncology research, continuously optimizing novel therapeutic strategies. Intratumor heterogeneity extensively consists of all tumor components, contributing to different tumor behaviors and treatment responses. Tumor-associated macrophages (TAMs), the core immune cells linking innate and adaptive immunity, play significant roles in tumor progression and resistance to therapies. Moreover, dynamic changes occur in TAM phenotypes and functions subject to the regulation of the tumor microenvironment. The heterogeneity of TAMs corresponding to the state of the tumor microenvironment has been comprehensively recognized using scRNA-seq. Herein, we reviewed recent research and summarized variations in TAM phenotypes and functions from a developmental perspective to better understand the significance of TAMs in the tumor microenvironment.
Collapse
Affiliation(s)
- Sen Yang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, and Peking Union Medical College, Beijing, 100730, China
| | - Mengyi Wang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, and Peking Union Medical College, Beijing, 100730, China
| | - Yuze Hua
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, and Peking Union Medical College, Beijing, 100730, China
| | - Jiayi Li
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, and Peking Union Medical College, Beijing, 100730, China
| | - Huaijin Zheng
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, and Peking Union Medical College, Beijing, 100730, China
| | - Ming Cui
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, and Peking Union Medical College, Beijing, 100730, China
| | - Nan Huang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, and Peking Union Medical College, Beijing, 100730, China
| | - Qiaofei Liu
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, and Peking Union Medical College, Beijing, 100730, China.
| | - Quan Liao
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
35
|
Brindle A, Bainbridge C, Kumar MR, Todryk S, Padget K. The Bisdioxopiperazine ICRF-193 Attenuates LPS-induced IL-1β Secretion by Macrophages. Inflammation 2024; 47:84-98. [PMID: 37656316 PMCID: PMC10798930 DOI: 10.1007/s10753-023-01895-2] [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: 05/26/2023] [Revised: 07/25/2023] [Accepted: 08/18/2023] [Indexed: 09/02/2023]
Abstract
Inhibiting pathological secretion of Interleukin-1β has shown beneficial effects in disease models and in the clinic and thus there is interest in finding inhibitors that can reduce its release from macrophages in response to their activation by foreign pathogens. We used an in vitro human macrophage model to investigate whether ICRF-193, a Topoisomerase II inhibitor could modulate IL1B mRNA expression and IL-1β secretion. These macrophage-like cells readily secrete IL-1β in response to Lipopolysaccharide (LPS). Upon exposure to a non-toxic dose of ICRF-193, IL-1β secretion was diminished by ~ 40%; however, level of transcription of IL1B was unaffected. We show that there was no Topoisomerase 2B (TOP2B) binding to several IL1B gene sites, which may explain why ICRF-193 does not alter IL1B mRNA levels. Hence, we show for the first time that ICRF-193 can reduce IL-1β secretion. Its low cost and the development of water-soluble prodrugs of ICRF-193 warrants its further investigation in the modulation of pathological secretion of this cytokine for the treatment of inflammatory disorders. (165 words).
Collapse
Affiliation(s)
- Ashleigh Brindle
- Faculty of Health and Life Sciences, Northumbria University at Newcastle, Newcastle Upon Tyne, NE1 8ST, UK
| | - Callum Bainbridge
- Faculty of Health and Life Sciences, Northumbria University at Newcastle, Newcastle Upon Tyne, NE1 8ST, UK
| | - Muganti R Kumar
- Faculty of Health and Life Sciences, Northumbria University at Newcastle, Newcastle Upon Tyne, NE1 8ST, UK
| | - Stephen Todryk
- Faculty of Health and Life Sciences, Northumbria University at Newcastle, Newcastle Upon Tyne, NE1 8ST, UK.
| | - Kay Padget
- Faculty of Health and Life Sciences, Northumbria University at Newcastle, Newcastle Upon Tyne, NE1 8ST, UK
| |
Collapse
|
36
|
Shaikh SS, Goebel A, Lee MC, Nahorski MS, Shenker N, Pamela Y, Drissi I, Brown C, Ison G, Shaikh MF, Kuttikat A, Woods WA, Dixit A, Stouffer K, Clarke MC, Menon DK, Woods CG. Evidence of a genetic background predisposing to complex regional pain syndrome type 1. J Med Genet 2024; 61:163-170. [PMID: 37816627 PMCID: PMC10850724 DOI: 10.1136/jmg-2023-109236] [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: 02/23/2023] [Accepted: 09/02/2023] [Indexed: 10/12/2023]
Abstract
BACKGROUND Complex regional pain syndrome type 1 (CRPS-1) is a rare, disabling and sometimes chronic disorder usually arising after a trauma. This exploratory study examined whether patients with chronic CRPS-1 have a different genetic profile compared with those who do not have the condition. METHODS Exome sequencing was performed to seek altered non-synonymous SNP allele frequencies in a discovery cohort of well-characterised patients with chronic CRPS-1 (n=34) compared with population databases. Identified SNP alleles were confirmed by Sanger sequencing and sought in a replication cohort (n=50). Gene expression of peripheral blood macrophages was assessed. RESULTS In the discovery cohort, the rare allele frequencies of four non-synonymous SNPs were statistically increased. The replication cohort confirmed this finding. In a chronic pain cohort, these alleles were not overexpressed. In total, 25 out of 84 (29.8%) patients with CRPS-1 expressed a rare allele. The SNPs were rs41289586 in ANO10, rs28360457 in P2RX7, rs1126930 in PRKAG1 and rs80308281 in SLC12A9. Males were more likely than females to have a rare SNP allele, 8 out of 14 (57.1%) vs 17 out of 70 (24.3%) (Fisher's p=0.023). ANO10, P2RX7, PRKAG1 and SLC12A9 were all expressed in macrophages from healthy human controls. CONCLUSION A single SNP in each of the genes ANO10, P2RX7, PRKAG1 and SLC12A9 was associated with developing chronic CRPS-1, with more males than females expressing these rare alleles. Our work suggests the possibility that a permissive genetic background is an important factor in the development of CRPS-1.
Collapse
Affiliation(s)
- Samiha S Shaikh
- Medical Genetics, Cambridge Institute for Medical Research, Cambridge, Cambridgeshire, UK
| | - Andreas Goebel
- Pain Research Institute, Clinical Sciences Centre, University of Liverpool Faculty of Health and Life Sciences, Liverpool, UK
| | - Michael C Lee
- Department of Medicine, Addenbrooke's Hospital, Cambridge, Cambridgeshire, UK
| | - Michael S Nahorski
- Medical Genetics, Cambridge Institute for Medical Research, Cambridge, Cambridgeshire, UK
| | - Nicholas Shenker
- Department of Rheumatology, Addenbrooke's Hospital Rheumatology Department, Cambridge, Cambridgeshire, UK
| | - Yunisa Pamela
- Medical Genetics, Cambridge Institute for Medical Research, Cambridge, Cambridgeshire, UK
- Department of Biomedical Sciences, Universitas Padjadjaran, Bandung, Indonesia
| | - Ichrak Drissi
- Medical Genetics, Cambridge Institute for Medical Research, Cambridge, Cambridgeshire, UK
| | - Christopher Brown
- Department of Medicine, Addenbrooke's Hospital, Cambridge, Cambridgeshire, UK
| | - Gillian Ison
- Department of Medicine, Addenbrooke's Hospital, Cambridge, Cambridgeshire, UK
| | - Maliha F Shaikh
- Department of Rheumatology, Addenbrooke's Hospital Rheumatology Department, Cambridge, Cambridgeshire, UK
| | - Anoop Kuttikat
- Department of Rheumatology, Addenbrooke's Hospital Rheumatology Department, Cambridge, Cambridgeshire, UK
| | - William A Woods
- Medical Genetics, Cambridge Institute for Medical Research, Cambridge, Cambridgeshire, UK
| | - Abhishek Dixit
- Department of Medicine, Addenbrooke's Hospital, Cambridge, Cambridgeshire, UK
| | - Kaitlin Stouffer
- Medical Genetics, Cambridge Institute for Medical Research, Cambridge, Cambridgeshire, UK
| | - Murray Ch Clarke
- Heart and Lung Research Institute, Cambridge Biomedical Campus, Cambridge, Cambridgeshire, UK
| | - David K Menon
- Brain Physics Laboratory, University of Cambridge Department of Clinical Neurosciences, Cambridge, Cambridgeshire, UK
| | - C Geoffrey Woods
- Medical Genetics, Cambridge Institute for Medical Research, Cambridge, Cambridgeshire, UK
| |
Collapse
|
37
|
Félix J, Martínez de Toda I, Díaz-Del Cerro E, Gil-Agudo F, De la Fuente M. The immunity and redox clocks in mice, markers of lifespan. Sci Rep 2024; 14:1703. [PMID: 38242936 PMCID: PMC10799057 DOI: 10.1038/s41598-024-51978-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 01/10/2024] [Indexed: 01/21/2024] Open
Abstract
Immune function and redox markers are used for estimating the aging rate, namely biological age (BA). However, it is unknown if this BA and its changes can be reflected in longevity. Thus, we must quantify BA in experimental animals. In peritoneal immune cells of 202 female mice (ICR/CD1) in different ages, 10 immune and 6 redox parameters were evaluated to construct two mathematical models for BA quantification in mice by multiple linear regression. Immune and redox parameters were selected as independent variables and chronological age as dependent, developing two models: the Immunity and the Redox Clocks, reaching both an adjusted R2 of 80.9% and a standard error of 6.38 and 8.57 weeks, respectively. Both models were validated in a different group of healthy mice obtaining a Pearson's correlation coefficient of 0.844 and 0.800 (p < 0.001) between chronological and BA. Furthermore, they were applied to adult prematurely aging mice, which showed a higher BA than non-prematurely aging mice. Moreover, after positive and negative lifestyle interventions, mice showed a lower and higher BA, respectively, than their age-matched controls. In conclusion, the Immunity and Redox Clocks allow BA quantification in mice and both the ImmunolAge and RedoxAge in mice relate to lifespan.
Collapse
Affiliation(s)
- Judith Félix
- Department of Genetics, Physiology and Microbiology (Animal Physiology Unit), Faculty of Biological Sciences, Complutense University of Madrid, 28040, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital, 12 de Octubre (imas12), 28041, Madrid, Spain
| | - Irene Martínez de Toda
- Department of Genetics, Physiology and Microbiology (Animal Physiology Unit), Faculty of Biological Sciences, Complutense University of Madrid, 28040, Madrid, Spain.
- Instituto de Investigación Sanitaria Hospital, 12 de Octubre (imas12), 28041, Madrid, Spain.
| | - Estefanía Díaz-Del Cerro
- Department of Genetics, Physiology and Microbiology (Animal Physiology Unit), Faculty of Biological Sciences, Complutense University of Madrid, 28040, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital, 12 de Octubre (imas12), 28041, Madrid, Spain
| | - Fernando Gil-Agudo
- Department of Genetics, Physiology and Microbiology (Animal Physiology Unit), Faculty of Biological Sciences, Complutense University of Madrid, 28040, Madrid, Spain
| | - Mónica De la Fuente
- Department of Genetics, Physiology and Microbiology (Animal Physiology Unit), Faculty of Biological Sciences, Complutense University of Madrid, 28040, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital, 12 de Octubre (imas12), 28041, Madrid, Spain
| |
Collapse
|
38
|
Şen B, Balcı‐Peynircioğlu B. Cellular models in autoinflammatory disease research. Clin Transl Immunology 2024; 13:e1481. [PMID: 38213819 PMCID: PMC10784111 DOI: 10.1002/cti2.1481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/12/2023] [Accepted: 12/12/2023] [Indexed: 01/13/2024] Open
Abstract
Systemic autoinflammatory diseases are a heterogeneous group of rare genetic disorders caused by dysregulation of the innate immune system. Understanding the complex mechanisms underlying these conditions is critical for developing effective treatments. Cellular models are essential for identifying new conditions and studying their pathogenesis. Traditionally, these studies have used primary cells and cell lines of disease-relevant cell types, although newer induced pluripotent stem cell (iPSC)-based models might have unique advantages. In this review, we discuss the three cellular models used in autoinflammatory disease research, their strengths and weaknesses, and their applications to inform future research in the field.
Collapse
Affiliation(s)
- Başak Şen
- Department of Medical BiologyHacettepe University Faculty of Medicine, SıhhiyeAnkaraTurkey
| | | |
Collapse
|
39
|
Selig M, Poehlman L, Lang NC, Völker M, Rolauffs B, Hart ML. Prediction of six macrophage phenotypes and their IL-10 content based on single-cell morphology using artificial intelligence. Front Immunol 2024; 14:1336393. [PMID: 38239351 PMCID: PMC10794337 DOI: 10.3389/fimmu.2023.1336393] [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: 11/10/2023] [Accepted: 12/14/2023] [Indexed: 01/22/2024] Open
Abstract
Introduction The last decade has led to rapid developments and increased usage of computational tools at the single-cell level. However, our knowledge remains limited in how extracellular cues alter quantitative macrophage morphology and how such morphological changes can be used to predict macrophage phenotype as well as cytokine content at the single-cell level. Methods Using an artificial intelligence (AI) based approach, this study determined whether (i) accurate macrophage classification and (ii) prediction of intracellular IL-10 at the single-cell level was possible, using only morphological features as predictors for AI. Using a quantitative panel of shape descriptors, our study assessed image-based original and synthetic single-cell data in two different datasets in which CD14+ monocyte-derived macrophages generated from human peripheral blood monocytes were initially primed with GM-CSF or M-CSF followed by polarization with specific stimuli in the presence/absence of continuous GM-CSF or M-CSF. Specifically, M0, M1 (GM-CSF-M1, TNFα/IFNγ-M1, GM-CSF/TNFα/IFNγ-M1) and M2 (M-CSF-M2, IL-4-M2a, M-CSF/IL-4-M2a, IL-10-M2c, M-CSF/IL-10-M2c) macrophages were examined. Results Phenotypes were confirmed by ELISA and immunostaining of CD markers. Variations of polarization techniques significantly changed multiple macrophage morphological features, demonstrating that macrophage morphology is a highly sensitive, dynamic marker of phenotype. Using original and synthetic single-cell data, cell morphology alone yielded an accuracy of 93% for the classification of 6 different human macrophage phenotypes (with continuous GM-CSF or M-CSF). A similarly high phenotype classification accuracy of 95% was reached with data generated with different stimuli (discontinuous GM-CSF or M-CSF) and measured at a different time point. These comparably high accuracies clearly validated the here chosen AI-based approach. Quantitative morphology also allowed prediction of intracellular IL-10 with 95% accuracy using only original data. Discussion Thus, image-based machine learning using morphology-based features not only (i) classified M0, M1 and M2 macrophages but also (ii) classified M2a and M2c subtypes and (iii) predicted intracellular IL-10 at the single-cell level among six phenotypes. This simple approach can be used as a general strategy not only for macrophage phenotyping but also for prediction of IL-10 content of any IL-10 producing cell, which can help improve our understanding of cytokine biology at the single-cell level.
Collapse
Affiliation(s)
- Mischa Selig
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
| | - Logan Poehlman
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
| | - Nils C Lang
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Marita Völker
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
| | - Bernd Rolauffs
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
| | - Melanie L Hart
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
40
|
Yan Y, Li Y, Chi Y, Ji M, Shen Y, Zou L. A comparative study of biological properties of three root canal sealers. Clin Oral Investig 2023; 28:11. [PMID: 38129367 DOI: 10.1007/s00784-023-05402-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023]
Abstract
OBJECTIVES The aim of this study was to evaluate the effects of Hiflow with other two kinds of root canal sealers on the biological behavior of stem cells from the apical papilla (SCAP), the influence on inflammatory cytokines release and its antibacterial effects. MATERIALS AND METHODS Material extracts of Hiflow, iRoot SP, and AH Plus were prepared. Then, SCAP was incubated with extracts. The effects were evaluated by CCK-8, wound healing assay, ALP staining, alizarin red staining, and qRT-PCR. Meanwhile, polymorphonuclears (PMNs) and monocytes were isolated and treated with extracts for 4 h and 24 h respectively. Cell viability was analyzed by Annexin-V/PI double staining flow cytometry. The effects on the release of cytokines were observed by ELISA. The antibacterial effects of different sealers were tested against three kinds of bacteria found in chronic apical periodontitis. RESULTS A series of results of SCAP showed that Hiflow and iRoot SP could promote cell proliferation, migration, and osteogenesis (p < 0.05). Although Hiflow was associated with greater cell apoptosis and necrosis when incubated with PMNs and monocytes (p < 0.05), it had an approximate release of anti-inflammatory cytokines with iRoot SP, which was higher than AH plus (p < 0.05). The co-culture showed that Hiflow and iRoot SP inhibited the colony formation of F. nucleatum (p < 0.05). However, both sealers had no obvious antibacterial effect on E. faecalis and P. gingivalis (p > 0.05). CONCLUSIONS In summary, Hiflow and iRoot SP both had positive biological stimulus on SCAP. Meanwhile, Hiflow showed a better induction on anti-inflammatory cytokines over the others. All the properties mentioned above and its antibacterial effect of F. nucleatum promise Hiflow a bright application prospect in endodontic uses. CLINICAL RELEVANCE References for clinical work to use BC Sealer Hiflow as a good biological root canal sealer.
Collapse
Affiliation(s)
- Yujia Yan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yanyao Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yaqi Chi
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Mengzhen Ji
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ya Shen
- Division of Endodontics, Department of Oral Biological & Medical Sciences, Faculty of Dentistry, The University of British Columbia, Vancouver, British Columbia, Canada.
| | - Ling Zou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
41
|
Zhang D, He J, Ding X, Wang R, Chen W. GENIPOSIDE IMPROVES CLP-INDUCED SEPSIS MODEL PROGNOSIS BY UPREGULATING PPARγ TO MODULATE MONOCYTE PHENOTYPE AND CYTOKINE NETWORK. Shock 2023; 60:753-761. [PMID: 37878499 DOI: 10.1097/shk.0000000000002239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2023]
Abstract
ABSTRACT Background : We explored the efficacy and main biological mechanism of geniposide intervention in sepsis. Methods : A sepsis model was established in male BALB/c mice through cecal ligation and puncture (CLP). Different doses of geniposide (20 or 40 mg/kg) were administered intravenously at 0 and/or 24 h after CLP surgery. The survival rate of different groups was observed. In addition, the expression levels of CD16 and major histocompatibility complex class II in monocytes were assessed using flow cytometry. The concentrations of TNF-α, IL-1β, IL-6, and IL-10 in the serum were measured by ELISA. We also observed the biological effects of geniposide on CD16 and MHC-II expression levels in RAW264.7 cells, as well as the secretion of TNF-α, IL-1β, IL-6, and IL-10 in the LPS-induced RAW264.7 cell model. The PPARγ levels were determined using western blot analysis. Results : Intravenous administration of 40 mg/kg of geniposide at 0 h after CLP significantly improved the survival outcomes in the septic mouse model, with no significant benefits from low dosing (20 mg/kg) or delayed administration (24 h). The effective dose of geniposide significantly decreased the serum cytokine TNF-α, IL-1β, IL-6, and IL-10 concentrations in septic mice ( P < 0.05). Notably, in vitro assays showed that geniposide specifically increased the IL-10 level. Geniposide significantly reduced the CD16 expression ( P < 0.05) and increased MHC-II expression in monocytes ( P < 0.05). In addition, geniposide elevated the PPARγ level in monocytes ( P < 0.05). Conclusions : High-dose early-stage geniposide administration significantly improved the survival rate in a CLP mouse sepsis model by modulating the monocyte phenotype and regulating the cytokine network (IL-6/IL-10 levels). The pharmacological mechanism of geniposide action might be exerted primarily through PPARγ upregulation.
Collapse
Affiliation(s)
- Dewen Zhang
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jian He
- Department of Emergency and Critical Care Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Xian Ding
- Department of Emergency and Critical Care Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Rui Wang
- Department of Emergency and Critical Care Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Wei Chen
- Department of Critical Care Medicine, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
42
|
Testa A, Quaglia F, Naranjo NM, Verrillo CE, Shields CD, Lin S, Pickles MW, Hamza DF, Von Schalscha T, Cheresh DA, Leiby B, Liu Q, Ding J, Kelly WK, Hooper DC, Corey E, Plow EF, Altieri DC, Languino LR. Targeting the αVβ3/NgR2 pathway in neuroendocrine prostate cancer. Matrix Biol 2023; 124:49-62. [PMID: 37956856 PMCID: PMC10823877 DOI: 10.1016/j.matbio.2023.11.003] [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: 07/18/2023] [Revised: 10/25/2023] [Accepted: 11/08/2023] [Indexed: 11/15/2023]
Abstract
Highly aggressive, metastatic, neuroendocrine prostate cancer, which typically develops from prostate cancer cells acquiring resistance to androgen deprivation therapy, is associated with limited treatment options and hence poor prognosis. We have previously demonstrated that the αVβ3 integrin is over-expressed in neuroendocrine prostate cancer. We now show that LM609, a monoclonal antibody that specifically targets the human αVβ3 integrin, hinders the growth of neuroendocrine prostate cancer patient-derived xenografts in vivo. Our group has recently identified a novel αVβ3 integrin binding partner, NgR2, responsible for regulating the expression of neuroendocrine markers and for inducing neuroendocrine differentiation in prostate cancer cells. Through in vitro functional assays, we here demonstrate that NgR2 is crucial in promoting cell adhesion to αVβ3 ligands. Moreover, we describe for the first time co-fractionation of αVβ3 integrin and NgR2 in small extracellular vesicles derived from metastatic prostate cancer patients' plasma. These prostate cancer patient-derived small extracellular vesicles have a functional impact on human monocytes, increasing their adhesion to fibronectin. The monocytes incubated with small extracellular vesicles do not show an associated change in conventional polarization marker expression and appear to be in an early stage that may be defined as "adhesion competent". Overall, these findings allow us to better understand integrin-directed signaling and cell-cell communication during cancer progression. Furthermore, our results pave the way for new diagnostic and therapeutic perspectives for patients affected by neuroendocrine prostate cancer.
Collapse
Affiliation(s)
- Anna Testa
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, United States; Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Fabio Quaglia
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, United States; Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Nicole M Naranjo
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, United States; Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Cecilia E Verrillo
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, United States; Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Christopher D Shields
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, United States; Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Stephen Lin
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, United States; Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Maxwell W Pickles
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, United States; Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Drini F Hamza
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, United States; Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Tami Von Schalscha
- Department of Pathology, Moores Cancer Center, and Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, United States
| | - David A Cheresh
- Department of Pathology, Moores Cancer Center, and Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, United States
| | - Benjamin Leiby
- Division of Biostatistics, Department of Pharmacology, Physiology, and Cancer Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Qin Liu
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA, United States
| | - Jianyi Ding
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA, United States
| | - William K Kelly
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, United States
| | - D Craig Hooper
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, United States; Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA, United States
| | - Edward F Plow
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Dario C Altieri
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, United States
| | - Lucia R Languino
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, United States; Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
43
|
Jolly A, Fernández B, Mundo SL, Elguezabal N. Modeling Paratuberculosis in Laboratory Animals, Cells, or Tissues: A Focus on Their Applications for Pathogenesis, Diagnosis, Vaccines, and Therapy Studies. Animals (Basel) 2023; 13:3553. [PMID: 38003170 PMCID: PMC10668694 DOI: 10.3390/ani13223553] [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: 08/12/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Paratuberculosis is a chronic granulomatous enteritis caused by Mycobacterium avium subsp. Paratuberculosis that affects a wide variety of domestic and wild animals. It is considered one of the diseases with the highest economic impact on the ruminant industry. Despite many efforts and intensive research, paratuberculosis control still remains controversial, and the existing diagnostic and immunoprophylactic tools have great limitations. Thus, models play a crucial role in understanding the pathogenesis of infection and disease, and in testing novel vaccine candidates. Ruminant animal models can be restricted by several reasons, related to space requirements, the cost of the animals, and the maintenance of the facilities. Therefore, we review the potential and limitations of the different experimental approaches currently used in paratuberculosis research, focusing on laboratory animals and cell-based models. The aim of this review is to offer a vision of the models that have been used, and what has been achieved or discovered with each one, so that the reader can choose the best model to answer their scientific questions and prove their hypotheses. Also, we bring forward new approaches that we consider worth exploring in the near future.
Collapse
Affiliation(s)
- Ana Jolly
- Cátedra de Inmunología, Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Av. Chorroarín 280, Buenos Aires C1427CWO, Argentina; (B.F.); (S.L.M.)
| | - Bárbara Fernández
- Cátedra de Inmunología, Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Av. Chorroarín 280, Buenos Aires C1427CWO, Argentina; (B.F.); (S.L.M.)
- Instituto de Investigaciones en Producción Animal (INPA), CONICET-Universidad de Buenos Aires, Av. Chorroarín 280, Buenos Aires C1427CWO, Argentina
- Instituto de Investigación y Tecnología en Reproducción Animal (INITRA), Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Av. Chorroarín 280, Buenos Aires C1427CWO, Argentina
| | - Silvia Leonor Mundo
- Cátedra de Inmunología, Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Av. Chorroarín 280, Buenos Aires C1427CWO, Argentina; (B.F.); (S.L.M.)
- Instituto de Investigaciones en Producción Animal (INPA), CONICET-Universidad de Buenos Aires, Av. Chorroarín 280, Buenos Aires C1427CWO, Argentina
- Instituto de Investigación y Tecnología en Reproducción Animal (INITRA), Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Av. Chorroarín 280, Buenos Aires C1427CWO, Argentina
| | - Natalia Elguezabal
- Departamento de Sanidad Animal, NEIKER-Instituto Vasco de Investigación y Desarrollo Agrario-Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain
| |
Collapse
|
44
|
Antonsen KW, Friis HN, Sorensen BS, Etzerodt A, Moestrup SK, Møller HJ. Comparison of culture media reveals that non-essential amino acids strongly affect the phenotype of human monocyte-derived macrophages. Immunology 2023; 170:344-358. [PMID: 37291897 DOI: 10.1111/imm.13670] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 05/20/2023] [Indexed: 06/10/2023] Open
Abstract
Macrophages are important innate immune cells with the ability to adapt their phenotype to environmental cues. Research on human macrophages often uses monocyte-derived macrophages cultured in vitro, but it is unclear if culture medium affects macrophage phenotype. The objective of this study was to determine the impact of culture medium composition on monocyte-derived macrophage phenotype. Monocyte-derived macrophages were generated in different formulations of culture media (RPMI 1640, DMEM, MEM, McCoy's 5a and IMDM). Viability, yield and cell size were monitored, and RT-qPCR, flow cytometry or ELISA was used to compare levels of phenotype markers (CD163, CD206, CD80, TNFα, IL-10, SIRPα, LILRB1 and Siglec-10). Yield, cell size, gene expression, membrane protein levels and release of soluble proteins were all affected by changes in culture medium composition. The most pronounced effects were observed after culture in DMEM, which lacks the non-essential amino acids asparagine, aspartic acid, glutamic acid and proline. Supplementation of DMEM with non-essential amino acids either fully or partly reversed most effects of DMEM on macrophage phenotype. The results suggest culture medium composition and amino acid availability affect the phenotype of human monocyte-derived macrophages cultured in vitro.
Collapse
Affiliation(s)
- Kristian W Antonsen
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Henriette N Friis
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Boe S Sorensen
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Anders Etzerodt
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Holger J Møller
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
45
|
Cao D, Saito S, Xu L, Fan W, Li X, Ahmed F, Jovanovic P, Shibata T, Che M, Bernstein EA, Gianni J, Divakaruni AS, Okwan-Duodu D, Khan Z, Riera CE, Chen F, Bernstein KE. Myeloid cell ACE shapes cellular metabolism and function in PCSK-9 induced atherosclerosis. Front Immunol 2023; 14:1278383. [PMID: 37928535 PMCID: PMC10623052 DOI: 10.3389/fimmu.2023.1278383] [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: 08/16/2023] [Accepted: 10/02/2023] [Indexed: 11/07/2023] Open
Abstract
The pathogenesis of atherosclerosis is defined by impaired lipid handling by macrophages which increases intracellular lipid accumulation. This dysregulation of macrophages triggers the accumulation of apoptotic cells and chronic inflammation which contributes to disease progression. We previously reported that mice with increased macrophage-specific angiotensin-converting enzyme, termed ACE10/10 mice, resist atherosclerosis in an adeno-associated virus-proprotein convertase subtilisin/kexin type 9 (AAV-PCSK9)-induced model. This is due to increased lipid metabolism by macrophages which contributes to plaque resolution. However, the importance of ACE in peripheral blood monocytes, which are the primary precursors of lesional-infiltrating macrophages, is still unknown in atherosclerosis. Here, we show that the ACE-mediated metabolic phenotype is already triggered in peripheral blood circulating monocytes and that this functional modification is directly transferred to differentiated macrophages in ACE10/10 mice. We found that Ly-6Clo monocytes were increased in atherosclerotic ACE10/10 mice. The monocytes isolated from atherosclerotic ACE10/10 mice showed enhanced lipid metabolism, elevated mitochondrial activity, and increased adenosine triphosphate (ATP) levels which implies that ACE overexpression is already altered in atherosclerosis. Furthermore, we observed increased oxygen consumption (VO2), respiratory exchange ratio (RER), and spontaneous physical activity in ACE10/10 mice compared to WT mice in atherosclerotic conditions, indicating enhanced systemic energy consumption. Thus, ACE overexpression in myeloid lineage cells modifies the metabolic function of peripheral blood circulating monocytes which differentiate to macrophages and protect against atherosclerotic lesion progression due to better lipid metabolism.
Collapse
Affiliation(s)
- DuoYao Cao
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Suguru Saito
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - LiMin Xu
- Department of Neurosurgery, Shenzhen Entry-Exit Frontier Inspection Hospital, Shenzhen, China
| | - Wei Fan
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Xiaomo Li
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Faizan Ahmed
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Predrag Jovanovic
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Tomohiro Shibata
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Mingtian Che
- Biobank and Pathology Shared Resource, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Ellen A. Bernstein
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Jorge Gianni
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Ajit S. Divakaruni
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA) David Geffen School of Medicine, Los Angeles, CA, United States
| | - Derick Okwan-Duodu
- Department of Pathology, Faculty of Medicine, Stanford University, San Jose, CA, United States
| | - Zakir Khan
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Celine E. Riera
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Fanfan Chen
- Department of Neurosurgery, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Kenneth E. Bernstein
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
46
|
Ahmad I, Naqvi RA, Valverde A, Naqvi AR. LncRNA MALAT1/microRNA-30b axis regulates macrophage polarization and function. Front Immunol 2023; 14:1214810. [PMID: 37860007 PMCID: PMC10582718 DOI: 10.3389/fimmu.2023.1214810] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 09/21/2023] [Indexed: 10/21/2023] Open
Abstract
Macrophages (Mφ) are long-lived myeloid cells that can polarize towards the proinflammatory M1 or proresolving M2 phenotype to control diverse biological processes such as inflammation, tissue damage, and regeneration. Noncoding RNA are a class of nonprotein-coding transcriptome with numerous interdependent biological roles; however, their functional interaction in the regulation of Mφ polarization and immune responses remain unclear. Here, we show antagonistic relationship between lncRNA (MALAT1) and microRNA (miR-30b) in shaping macrophage polarization and immune functions. MALAT1 expression displays a time-dependent induction during Mφ differentiation and, upon challenge with TLR4 agonist (E. coli LPS). MALAT1 knockdown promoted the expression of M2Mφ markers without affecting M1Mφ markers, suggesting that MALAT1 favors the M1 phenotype by suppressing M2 differentiation. Compared to the control, MALAT1 knockdown resulted in reduced antigen uptake and processing, bacterial phagocytosis, and bactericidal activity, strongly supporting its critical role in regulating innate immune functions in Mφ. Consistent with this, MALAT1 knockdown showed impaired cytokine secretion upon challenge with LPS. Importantly, MALAT1 exhibit an antagonistic expression pattern with all five members of the miR-30 family during M2 Mφ differentiation. Dual-luciferase assays validated a novel sequence on MALAT1 that interacts with miR-30b, a microRNA that promotes the M2 phenotype. Phagocytosis and antigen processing assays unequivocally demonstrated that MALAT1 and miR-30b are functionally antagonistic. Concurrent MALAT1 knockdown and miR-30b overexpression exhibited the most significant attenuation in both assays. In human subjects with periodontal disease and murine model of ligature-induced periodontitis, we observed higher levels of MALAT1, M1Mφ markers and downregulation of miR-30b expression in gingival tissues suggesting a pro-inflammatory function of MALAT1 in vivo. Overall, we unraveled the role of MALAT1 in Mφ polarization and delineated the underlying mechanism of its regulation by involving MALAT-1-driven miR-30b sequestration.
Collapse
Affiliation(s)
| | | | | | - Afsar R. Naqvi
- Mucosal Immunology Lab, College of Dentistry, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
47
|
Gorodilova AV, Kitaeva KV, Filin IY, Mayasin YP, Kharisova CB, Issa SS, Solovyeva VV, Rizvanov AA. The Potential of Dendritic Cell Subsets in the Development of Personalized Immunotherapy for Cancer Treatment. Curr Issues Mol Biol 2023; 45:8053-8070. [PMID: 37886952 PMCID: PMC10605421 DOI: 10.3390/cimb45100509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/27/2023] [Accepted: 09/30/2023] [Indexed: 10/28/2023] Open
Abstract
Since the discovery of dendritic cells (DCs) in 1973 by Ralph Steinman, a tremendous amount of knowledge regarding these innate immunity cells has been accumulating. Their role in regulating both innate and adaptive immune processes is gradually being uncovered. DCs are proficient antigen-presenting cells capable of activating naive T-lymphocytes to initiate and generate effective anti-tumor responses. Although DC-based immunotherapy has not yielded significant results, the substantial number of ongoing clinical trials underscores the relevance of DC vaccines, particularly as adjunctive therapy or in combination with other treatment options. This review presents an overview of current knowledge regarding human DCs, their classification, and the functions of distinct DC populations. The stepwise process of developing therapeutic DC vaccines to treat oncological diseases is discussed, along with speculation on the potential of combined therapy approaches and the role of DC vaccines in modern immunotherapy.
Collapse
Affiliation(s)
- Anna Valerevna Gorodilova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.V.G.); (K.V.K.); (I.Y.F.); (Y.P.M.); (C.B.K.); (V.V.S.)
| | - Kristina Viktorovna Kitaeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.V.G.); (K.V.K.); (I.Y.F.); (Y.P.M.); (C.B.K.); (V.V.S.)
| | - Ivan Yurevich Filin
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.V.G.); (K.V.K.); (I.Y.F.); (Y.P.M.); (C.B.K.); (V.V.S.)
| | - Yuri Pavlovich Mayasin
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.V.G.); (K.V.K.); (I.Y.F.); (Y.P.M.); (C.B.K.); (V.V.S.)
| | - Chulpan Bulatovna Kharisova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.V.G.); (K.V.K.); (I.Y.F.); (Y.P.M.); (C.B.K.); (V.V.S.)
| | - Shaza S. Issa
- Department of Genetics and Biotechnology, St. Petersburg State University, 199034 St. Petersburg, Russia;
| | - Valeriya Vladimirovna Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.V.G.); (K.V.K.); (I.Y.F.); (Y.P.M.); (C.B.K.); (V.V.S.)
| | - Albert Anatolyevich Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.V.G.); (K.V.K.); (I.Y.F.); (Y.P.M.); (C.B.K.); (V.V.S.)
| |
Collapse
|
48
|
Urakami S, Koma YI, Tsukamoto S, Azumi Y, Miyako S, Kitamura Y, Kodama T, Nishio M, Shigeoka M, Abe H, Usami Y, Kodama Y, Yokozaki H. Biological and clinical significance of the YKL-40/osteopontin-integrin β4-p70S6K axis induced by macrophages in early oesophageal squamous cell carcinoma. J Pathol 2023; 261:55-70. [PMID: 37436683 DOI: 10.1002/path.6148] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 05/12/2023] [Accepted: 05/27/2023] [Indexed: 07/13/2023]
Abstract
M2 macrophages contribute to the progression of oesophageal squamous cell carcinoma (ESCC); however, the roles of M2 macrophages in early ESCC remain unclear. To clarify the biological mechanisms underlying the interaction between M2 macrophages and oesophageal epithelial cells in early-stage ESCC, in vitro co-culture assays between the immortalised oesophageal epithelial cell line Het-1A and cytokine-defined M2 macrophages were established. Co-culture with M2 macrophages promoted the proliferation and migration of Het-1A cells via the mTOR-p70S6K signalling pathway activated by YKL-40, also known as chitinase 3-like 1, and osteopontin (OPN) that were hypersecreted in the co-culture supernatants. YKL-40 and OPN promoted the above phenotypes of Het-1A by making a complex with integrin β4 (β4). Furthermore, YKL-40 and OPN promoted M2 polarisation, proliferation, and migration of macrophages. To validate the pathological and clinical significances of in vitro experimental results, immunohistochemistry of human early ESCC tissues obtained by endoscopic submucosal dissection (ESD) was performed, confirming the activation of the YKL-40/OPN-β4-p70S6K axis in the tumour area. Moreover, epithelial expression of β4 and the number of epithelial and stromal infiltrating YKL-40- and OPN-positive cells correlated with the Lugol-voiding lesions (LVLs), a well-known predictor of the incidence of metachronous ESCC. Furthermore, the combination of high expression of β4 and LVLs or high numbers of epithelial and stromal infiltrating YKL-40- and OPN-positive immune cells could more clearly detect the incidence of metachronous ESCC than each of the parameters alone. Our results demonstrated that the YKL-40/OPN-β4-p70S6K axis played important roles in early-stage ESCC, and the high expression levels of β4 and high numbers of infiltrating YKL-40- and OPN-positive immune cells could be useful predictive parameters for the incidence of metachronous ESCC after ESD. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Satoshi Urakami
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yu-Ichiro Koma
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shuichi Tsukamoto
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuki Azumi
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
- Division of Gastro-intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shoji Miyako
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
- Division of Gastro-intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yu Kitamura
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
- Division of Gastro-intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takayuki Kodama
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Mari Nishio
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Manabu Shigeoka
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hirofumi Abe
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yu Usami
- Department of Oral and Maxillofacial Pathology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Yuzo Kodama
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiroshi Yokozaki
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
49
|
Pirttiniemi A, Adeshara K, Happonen N, Einarsdottir E, Katayama S, Salmenkari H, Hörkkö S, Kere J, Groop PH, Lehto M. Long-chain polyphosphates inhibit type I interferon signaling and augment LPS-induced cytokine secretion in human leukocytes. J Leukoc Biol 2023; 114:250-265. [PMID: 37224571 DOI: 10.1093/jleuko/qiad058] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 04/20/2023] [Accepted: 05/12/2023] [Indexed: 05/26/2023] Open
Abstract
Inorganic polyphosphates are evolutionarily conserved bioactive phosphate polymers found as various chain lengths in all living organisms. In mammals, polyphosphates play a vital role in the regulation of cellular metabolism, coagulation, and inflammation. Long-chain polyphosphates are found along with endotoxins in pathogenic gram-negative bacteria and can participate in bacterial virulence. We aimed to investigate whether exogenously administered polyphosphates modulate human leukocyte function in vitro by treating the cells with 3 different chain lengths of polyphosphates (P14, P100, and P700). The long-chain polyphosphates, P700, had a remarkable capacity to downregulate type I interferon signaling dose dependently in THP1-Dual cells while only a slight elevation could be observed in the NF-κB pathway with the highest dose of P700. P700 treatment decreased lipopolysaccharide-induced IFNβ transcription and secretion, reduced STAT1 phosphorylation, and downregulated subsequent interferon-stimulated gene expression in primary human peripheral blood mononuclear cells. P700 also augmented lipopolysaccharide-induced secretion of IL-1α, IL-1β, IL-4, IL-5, IL-10, and IFNγ. Furthermore, P700 has previously been reported to increase the phosphorylation of several intracellular signaling mediators, such as AKT, mTOR, ERK, p38, GSK3α/β, HSP27, and JNK pathway components, which was supported by our findings. Taken together, these observations demonstrate the extensive modulatory effects P700 has on cytokine signaling and the inhibitory effects specifically targeted to type I interferon signaling in human leukocytes.
Collapse
Affiliation(s)
- Anniina Pirttiniemi
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum, Haartmaninkatu 8, 00290 Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 4, 00290 Helsinki, Finland
- Clinical and Molecular Metabolism, Faculty of Medicine Research Programs, University of Helsinki, Biomedicum, Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Krishna Adeshara
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum, Haartmaninkatu 8, 00290 Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 4, 00290 Helsinki, Finland
- Clinical and Molecular Metabolism, Faculty of Medicine Research Programs, University of Helsinki, Biomedicum, Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Natalie Happonen
- Medical Microbiology and Immunology, Research Unit of Biomedicine, University of Oulu, Aapistie 5A, 90220 Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Aapistie 5A, 90220 Oulu, Finland
- Nordlab, Oulu University Hospital, Kajaanintie 50, 90220 Oulu, Finland
| | - Elisabet Einarsdottir
- Science for Life Laboratory, Department of Gene Technology, KTH-Royal Institute of Technology, Tomtebodavägen 23A, 17165 Solna, Sweden
| | - Shintaro Katayama
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum, Haartmaninkatu 8, 00290 Helsinki, Finland
- Stem Cells and Metabolism Research Program, University of Helsinki, Biomedicum, Haartmaninkatu 8, 00290 Helsinki, Finland
- Department of Biosciences and Nutrition, Karolinska Institutet, Neo, Blickagången 16, Flemingsberg, SE-14183 Huddinge, Sweden
| | - Hanne Salmenkari
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum, Haartmaninkatu 8, 00290 Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 4, 00290 Helsinki, Finland
- Clinical and Molecular Metabolism, Faculty of Medicine Research Programs, University of Helsinki, Biomedicum, Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Sohvi Hörkkö
- Medical Microbiology and Immunology, Research Unit of Biomedicine, University of Oulu, Aapistie 5A, 90220 Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Aapistie 5A, 90220 Oulu, Finland
| | - Juha Kere
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum, Haartmaninkatu 8, 00290 Helsinki, Finland
- Stem Cells and Metabolism Research Program, University of Helsinki, Biomedicum, Haartmaninkatu 8, 00290 Helsinki, Finland
- Department of Biosciences and Nutrition, Karolinska Institutet, Neo, Blickagången 16, Flemingsberg, SE-14183 Huddinge, Sweden
| | - Per-Henrik Groop
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum, Haartmaninkatu 8, 00290 Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 4, 00290 Helsinki, Finland
- Clinical and Molecular Metabolism, Faculty of Medicine Research Programs, University of Helsinki, Biomedicum, Haartmaninkatu 8, 00290 Helsinki, Finland
- Department of Diabetes, Central Clinical School, Monash University, Alfred Centre, 99 Commercial Road, Melbourne 3004, VIC, Australia
| | - Markku Lehto
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum, Haartmaninkatu 8, 00290 Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 4, 00290 Helsinki, Finland
- Clinical and Molecular Metabolism, Faculty of Medicine Research Programs, University of Helsinki, Biomedicum, Haartmaninkatu 8, 00290 Helsinki, Finland
| |
Collapse
|
50
|
Schrodt MV, Behan-Bush RM, Liszewski JN, Humpal-Pash ME, Boland LK, Scroggins SM, Santillan DA, Ankrum JA. Efferocytosis of viable versus heat-inactivated MSC induces human monocytes to distinct immunosuppressive phenotypes. Stem Cell Res Ther 2023; 14:206. [PMID: 37592321 PMCID: PMC10433682 DOI: 10.1186/s13287-023-03443-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 08/07/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND Immunomodulation by mesenchymal stromal cells (MSCs) can occur through trophic factor mechanisms, however, intravenously infused MSCs are rapidly cleared from the body yet a potent immunotherapeutic response is still observed. Recent work suggests that monocytes contribute to the clearance of MSCs via efferocytosis, the body's natural mechanism for clearing dead and dying cells in a non-inflammatory manner. This begs the questions of how variations in MSC quality affect monocyte phenotype and if viable MSCs are even needed to elicit an immunosuppressive response. METHODS Herein, we sought to dissect MSC's trophic mechanism from their efferocytic mechanisms and determine if the viability of MSCs prior to efferocytosis influences the resultant phenotype of monocytes. We cultured viable or heat-inactivated human umbilical cord MSCs with human peripheral blood mononuclear cells for 24 h and observed changes in monocyte surface marker expression and secretion profile. To isolate the effect of efferocytosis from MSC trophic factors, we used cell separation techniques to remove non-efferocytosed MSCs before challenging monocytes to suppress T-cells or respond to inflammatory stimuli. For all experiments, viable and heat-inactivated efferocytic-licensing of monocytes were compared to non-efferocytic-licensing control. RESULTS We found that monocytes efferocytose viable and heat-inactivated MSCs equally, but only viable MSC-licensed monocytes suppress activated T-cells and suppression occurred even after depletion of residual MSCs. This provides direct evidence that monocytes that efferocytose viable MSCs are immunosuppressive. Further characterization of monocytes after efferocytosis showed that uptake of viable-but not heat inactivated-MSC resulted in monocytes secreting IL-10 and producing kynurenine. When monocytes were challenged with LPS, IL-2, and IFN-γ to simulate sepsis, monocytes that had efferocytosed viable MSC had higher levels of IDO while monocytes that efferocytosed heat inactivated-MSCs produced the lowest levels of TNF-α. CONCLUSION Collectively, these studies show that the quality of MSCs efferocytosed by monocytes polarize monocytes toward distinctive immunosuppressive phenotypes and highlights the need to tailor MSC therapies for specific indications.
Collapse
Affiliation(s)
- Michael V Schrodt
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52245, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52245, USA
| | - Riley M Behan-Bush
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52245, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52245, USA
| | - Jesse N Liszewski
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52245, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52245, USA
| | - Madeleine E Humpal-Pash
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52245, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52245, USA
| | - Lauren K Boland
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52245, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52245, USA
| | - Sabrina M Scroggins
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Center for Immunology and Immune Based Diseases, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Biomedical Sciences, Center for Immunology, Center for Clinical and Translational Science, University of Minnesota School of Medicine, Duluth, MN, USA
| | - Donna A Santillan
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Center for Immunology and Immune Based Diseases, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Biomedical Sciences, Center for Immunology, Center for Clinical and Translational Science, University of Minnesota School of Medicine, Duluth, MN, USA
| | - James A Ankrum
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52245, USA.
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52245, USA.
- , 103 S. Capitol St., 5621 SC, Iowa City, IA, 52242, USA.
| |
Collapse
|