1
|
Datta M. Analysis of Germinal Center Reaction in Competitive Bone Marrow Chimeric Rag2 -/-γc -/-Mice. Methods Mol Biol 2025; 2909:19-29. [PMID: 40029512 DOI: 10.1007/978-1-0716-4442-3_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] [Indexed: 03/05/2025]
Abstract
Germinal center (GC) reaction is crucial for the generation of high-affinity antibodies against any infection. To address the role of specific genes in GC, knockout mouse models are generally used. However, since GC is a multicellular event, complete knockout models cannot pinpoint the cell-intrinsic effect of certain genes in GC reaction. Here, we describe a detailed protocol for the analysis of GC in competitive bone marrow (BM) chimeric mice generated by transplanting 1:1 mixture of CD45.1 and CD45.2 positive donor bone marrow (BM) cells in immunodeficient Rag2-/-γc-/- mice as recipient host. We describe the method of immunization using sheep red blood cells (SRBC), detection of successful immunization by the antigen-specific antibody titer in the serum of the immunized mice and finally assessing the GC reaction in the spleen of the immunized mice by flow cytometry 10 days post-immunization. When the competitive chimera is made with wild type (WT) and a knockout (KO) donor BM, this method is suitable to address the cell type-specific (e.g., B-cell specific) role of the KO gene in GC.
Collapse
Affiliation(s)
- Moumita Datta
- Institute of Immunology, Ulm University Medical Center, Ulm, Germany.
| |
Collapse
|
2
|
Hasanali ZS, Garfall AL, Burzenski L, Shultz LD, Tang Y, Kadu S, Sheppard NC, Liu W, Dopkin D, Vogl DT, Cohen AD, Waxman AJ, Susanibar-Adaniya SP, Carroll M, Stadtmauer EA, Allman D. Human IL-6 fosters long-term engraftment of patient-derived disease-driving myeloma cells in immunodeficient mice. JCI Insight 2024; 9:e177300. [PMID: 38713510 PMCID: PMC11141932 DOI: 10.1172/jci.insight.177300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 04/17/2024] [Indexed: 05/09/2024] Open
Abstract
Multiple myeloma is a largely incurable and life-threatening malignancy of antibody-secreting plasma cells. An effective and widely available animal model that recapitulates human myeloma and related plasma cell disorders is lacking. We show that busulfan-conditioned human IL-6-transgenic (hIL-6-transgenic) NSG (NSG+hIL6) mice reliably support the engraftment of malignant and premalignant human plasma cells, including from patients diagnosed with monoclonal gammopathy of undetermined significance, pre- and postrelapse myeloma, plasma cell leukemia, and amyloid light chain amyloidosis. Consistent with human disease, NSG+hIL6 mice engrafted with patient-derived myeloma cells developed serum M spikes, and a majority developed anemia, hypercalcemia, and/or bone lesions. Single-cell RNA sequencing showed nonmalignant and malignant cell engraftment, the latter expressing a wide array of mRNAs associated with myeloma cell survival and proliferation. Myeloma-engrafted mice given CAR T cells targeting plasma cells or bortezomib experienced reduced tumor burden. Our results establish NSG+hIL6 mice as an effective patient-derived xenograft model for study and preclinical drug development of multiple myeloma and related plasma cell disorders.
Collapse
Affiliation(s)
- Zainul S. Hasanali
- Division of Hematology Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alfred L. Garfall
- Division of Hematology Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | - Yan Tang
- Center for Cellular Immunotherapies
| | | | - Neil C. Sheppard
- Center for Cellular Immunotherapies
- Department of Pathology and Laboratory Medicine, and
| | - Wei Liu
- Center for Cellular Immunotherapies
| | - Derek Dopkin
- Stem Cell and Xenograft Core Facility, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dan T. Vogl
- Division of Hematology Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Adam D. Cohen
- Division of Hematology Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Adam J. Waxman
- Division of Hematology Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Martin Carroll
- Division of Hematology Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Stem Cell and Xenograft Core Facility, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Edward A. Stadtmauer
- Division of Hematology Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David Allman
- Department of Pathology and Laboratory Medicine, and
| |
Collapse
|
3
|
Dettmer-Monaco V, Weißert K, Ammann S, Monaco G, Lei L, Gräßel L, Rhiel M, Rositzka J, Kaufmann MM, Geiger K, Andrieux G, Lao J, Thoulass G, Schell C, Boerries M, Illert AL, Cornu TI, Ehl S, Aichele P, Cathomen T. Gene editing of hematopoietic stem cells restores T-cell response in familial hemophagocytic lymphohistiocytosis. J Allergy Clin Immunol 2024; 153:243-255.e14. [PMID: 37595758 DOI: 10.1016/j.jaci.2023.08.003] [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: 05/07/2023] [Revised: 07/04/2023] [Accepted: 08/08/2023] [Indexed: 08/20/2023]
Abstract
BACKGROUND Hemophagocytic lymphohistiocytosis (HLH) is a hyperinflammatory disorder characterized by a life-threatening cytokine storm and immunopathology. Familial HLH type 3 (FHL3) accounts for approximately 30% of all inborn HLH cases worldwide. It is caused by mutations in the UNC13D gene that result in impaired degranulation of cytotoxic vesicles and hence compromised T-cell- and natural killer-cell-mediated killing. Current treatment protocols, including allogeneic hematopoietic stem cell (HSC) transplantation, still show high mortality. OBJECTIVE We sought to develop and evaluate a curative genome editing strategy in the preclinical FHL3 Jinx mouse model. Jinx mice harbor a cryptic splice donor site in Unc13d intron 26 and develop clinical symptoms of human FHL3 upon infection with lymphocytic choriomeningitis virus (LCMV). METHODS We employed clustered regularly interspaced short palindromic repeats (CRISPR)-Cas technology to delete the disease-causing mutation in HSCs and transplanted Unc13d-edited stem cells into busulfan-conditioned Jinx recipient mice. Safety studies included extensive genotyping and chromosomal aberrations analysis by single targeted linker-mediated PCR sequencing (CAST-Seq)-based off-target analyses. Cure from HLH predisposition was assessed by LCMV infection. RESULTS Hematopoietic cells isolated from transplanted mice revealed efficient gene editing (>95%), polyclonality of the T-cell receptor repertoire, and neither signs of off-target effects nor leukemogenesis. Unc13d transcription levels of edited and wild-type cells were comparable. While LCMV challenge resulted in acute HLH in Jinx mice transplanted with mock-edited HSCs, Jinx mice grafted with Unc13d-edited cells showed rapid virus clearance and protection from HLH. CONCLUSIONS Our study demonstrates that transplantation of CRISPR-Cas edited HSCs supports the development of a functional polyclonal T-cell response in the absence of genotoxicity-associated clonal outgrowth.
Collapse
Affiliation(s)
- Viviane Dettmer-Monaco
- Institute for Transfusion Medicine and Gene Therapy, Medical Center-University of Freiburg, Freiburg; Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Freiburg
| | - Kristoffer Weißert
- Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Freiburg; Institute for Immunodeficiency, Medical Center-University of Freiburg, Freiburg
| | - Sandra Ammann
- Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Freiburg; Institute for Immunodeficiency, Medical Center-University of Freiburg, Freiburg
| | - Gianni Monaco
- Institute for Transfusion Medicine and Gene Therapy, Medical Center-University of Freiburg, Freiburg; Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Freiburg; Institute of Neuropathology, Medical Center-University of Freiburg, Freiburg
| | - Lei Lei
- Institute for Transfusion Medicine and Gene Therapy, Medical Center-University of Freiburg, Freiburg; Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Freiburg; Ph.D. Program, Faculty of Biology, University of Freiburg, Freiburg
| | - Linda Gräßel
- Department of Internal Medicine I, Medical Center-University of Freiburg, Freiburg; German Cancer Consortium, Partner Site Freiburg & German Cancer Research Center, Heidelberg
| | - Manuel Rhiel
- Institute for Transfusion Medicine and Gene Therapy, Medical Center-University of Freiburg, Freiburg; Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Freiburg
| | - Julia Rositzka
- Institute for Transfusion Medicine and Gene Therapy, Medical Center-University of Freiburg, Freiburg; Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Freiburg
| | - Masako M Kaufmann
- Institute for Transfusion Medicine and Gene Therapy, Medical Center-University of Freiburg, Freiburg; Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Freiburg; Spemann Graduate School of Biology and Medicine, University of Freiburg, Freiburg
| | - Kerstin Geiger
- Institute for Transfusion Medicine and Gene Therapy, Medical Center-University of Freiburg, Freiburg; Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Freiburg; Ph.D. Program, Faculty of Biology, University of Freiburg, Freiburg
| | - Geoffroy Andrieux
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Freiburg; Faculty of Medicine, University of Freiburg, Freiburg
| | - Jessica Lao
- Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Freiburg; Institute for Immunodeficiency, Medical Center-University of Freiburg, Freiburg; Ph.D. Program, Faculty of Biology, University of Freiburg, Freiburg
| | - Gudrun Thoulass
- Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Freiburg; Institute for Immunodeficiency, Medical Center-University of Freiburg, Freiburg; Ph.D. Program, Faculty of Biology, University of Freiburg, Freiburg
| | - Christoph Schell
- Faculty of Medicine, University of Freiburg, Freiburg; Institute of Surgical Pathology, Medical Center-University of Freiburg, Freiburg
| | - Melanie Boerries
- German Cancer Consortium, Partner Site Freiburg & German Cancer Research Center, Heidelberg; Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Freiburg; Faculty of Medicine, University of Freiburg, Freiburg
| | - Anna L Illert
- Department of Internal Medicine I, Medical Center-University of Freiburg, Freiburg; German Cancer Consortium, Partner Site Freiburg & German Cancer Research Center, Heidelberg; Faculty of Medicine, University of Freiburg, Freiburg
| | - Tatjana I Cornu
- Institute for Transfusion Medicine and Gene Therapy, Medical Center-University of Freiburg, Freiburg; Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Freiburg; Faculty of Medicine, University of Freiburg, Freiburg
| | - Stephan Ehl
- Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Freiburg; Institute for Immunodeficiency, Medical Center-University of Freiburg, Freiburg; Faculty of Medicine, University of Freiburg, Freiburg
| | - Peter Aichele
- Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Freiburg; Institute for Immunodeficiency, Medical Center-University of Freiburg, Freiburg; Faculty of Medicine, University of Freiburg, Freiburg
| | - Toni Cathomen
- Institute for Transfusion Medicine and Gene Therapy, Medical Center-University of Freiburg, Freiburg; Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Freiburg; Faculty of Medicine, University of Freiburg, Freiburg.
| |
Collapse
|
4
|
Phu TA, Vu NK, Ng M, Gao AS, Stoolman JS, Chandel NS, Raffai RL. ApoE enhances mitochondrial metabolism via microRNA-142a/146a-regulated circuits that suppress hematopoiesis and inflammation in hyperlipidemia. Cell Rep 2023; 42:113206. [PMID: 37824329 DOI: 10.1016/j.celrep.2023.113206] [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: 03/21/2022] [Revised: 05/08/2023] [Accepted: 09/19/2023] [Indexed: 10/14/2023] Open
Abstract
Apolipoprotein E (ApoE) is recognized for its pleiotropic properties that suppress inflammation. We report that ApoE serves as a metabolic rheostat that regulates microRNA control of glycolytic and mitochondrial activity in myeloid cells and hematopoietic stem and progenitor cells (HSPCs). ApoE expression in myeloid cells increases microRNA-146a, which reduces nuclear factor κB (NF-κB)-driven GLUT1 expression and glycolytic activity. In contrast, ApoE expression reduces microRNA-142a, which increases carnitine palmitoyltransferase 1a (CPT1A) expression, fatty acid oxidation, and oxidative phosphorylation. Improved mitochondrial metabolism by ApoE expression causes an enrichment of tricarboxylic acid (TCA) cycle metabolites and nicotinamide adenine dinucleotide (NAD+) in macrophages. The study of mice with conditional ApoE expression supports the capacity of ApoE to foster microRNA-controlled immunometabolism. Modulation of microRNA-146a and -142a in the hematopoietic system of hyperlipidemic mice using RNA mimics and antagonists, respectively, improves mitochondrial metabolism, which suppresses inflammation and hematopoiesis. Our findings unveil microRNA regulatory circuits, controlled by ApoE, that exert metabolic control over hematopoiesis and inflammation in hyperlipidemia.
Collapse
Affiliation(s)
- Tuan Anh Phu
- Department of Veterans Affairs, Surgical Service (112G), San Francisco VA Medical Center, San Francisco, CA 94121, USA; Northern California Institute for Research and Education, San Francisco, CA 94121, USA
| | - Ngan K Vu
- Department of Veterans Affairs, Surgical Service (112G), San Francisco VA Medical Center, San Francisco, CA 94121, USA; Northern California Institute for Research and Education, San Francisco, CA 94121, USA
| | - Martin Ng
- Department of Veterans Affairs, Surgical Service (112G), San Francisco VA Medical Center, San Francisco, CA 94121, USA; Northern California Institute for Research and Education, San Francisco, CA 94121, USA
| | - Alex S Gao
- Department of Veterans Affairs, Surgical Service (112G), San Francisco VA Medical Center, San Francisco, CA 94121, USA; Northern California Institute for Research and Education, San Francisco, CA 94121, USA
| | - Joshua S Stoolman
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Navdeep S Chandel
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Department of Biochemistry & Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Robert L Raffai
- Department of Veterans Affairs, Surgical Service (112G), San Francisco VA Medical Center, San Francisco, CA 94121, USA; Northern California Institute for Research and Education, San Francisco, CA 94121, USA; Department of Surgery, Division of Endovascular and Vascular Surgery, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
5
|
Maidana DE, Gonzalez-Buendia L, Miller JW, Vavvas DG. RIPK necrotic cell death pathway in both donor photoreceptor and host immune cells synergize to affect photoreceptor graft survival. FASEB J 2023; 37:e22847. [PMID: 36862516 PMCID: PMC10590064 DOI: 10.1096/fj.202201137r] [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: 07/16/2022] [Revised: 11/29/2022] [Accepted: 02/15/2023] [Indexed: 03/03/2023]
Abstract
Photoreceptor transplant has been put forward as a repair strategy to tackle degenerated retinas. Nonetheless, cell death and immune rejection seriously limit the success of this strategy, with only a small fraction of transplanted cells surviving. Improving the survival of transplanted cells is of critical importance. Recent evidence has identified receptor-interacting protein kinase 3 (RIPK3) as a molecular trigger controlling necroptotic cell death and inflammation. However, its role in photoreceptor transplantation and regenerative medicine has not been studied. We hypothesized that modulation of RIPK3 to address both cell death and immunity could have advantageous effects on photoreceptor survival. In a model of inherited retinal degeneration, deletion of RIPK3 in donor photoreceptor precursors significantly increases the survival of transplanted cells. Simultaneous RIPK3 deletion in donor photoreceptors and recipients maximizes graft survival. Lastly, to discern the role of RIPK3 in the host immune response, bone marrow transplant experiments demonstrated that peripheral immune cell RIPK3 deficiency is protective for both donor and host photoreceptor survival. Interestingly, this finding is independent of photoreceptor transplantation, as the peripheral protective effect is also observed in an additional retinal detachment photoreceptor degeneration model. Altogether, these results indicate that immunomodulatory and neuroprotective strategies targeting the RIPK3 pathway can aid regenerative therapies of photoreceptor transplantation.
Collapse
Affiliation(s)
- Daniel E. Maidana
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Lucia Gonzalez-Buendia
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| | - Joan W. Miller
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| | - Demetrios G. Vavvas
- Retina Service, Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
6
|
Murillo-León M, Bastidas-Quintero AM, Endres NS, Schnepf D, Delgado-Betancourt E, Ohnemus A, Taylor GA, Schwemmle M, Staeheli P, Steinfeldt T. IFN-λ is protective against lethal oral Toxoplasma gondii infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.24.529861. [PMID: 36865100 PMCID: PMC9980175 DOI: 10.1101/2023.02.24.529861] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
Abstract
Interferons are essential for innate and adaptive immune responses against a wide variety of pathogens. Interferon lambda (IFN-λ) protects mucosal barriers during pathogen exposure. The intestinal epithelium is the first contact site for Toxoplasma gondii (T. gondii) with its hosts and the first defense line that limits parasite infection. Knowledge of very early T. gondii infection events in the gut tissue is limited and a possible contribution of IFN-λ has not been investigated so far. Here, we demonstrate with systemic interferon lambda receptor (IFNLR1) and conditional (Villin-Cre) knockout mouse models and bone marrow chimeras of oral T. gondii infection and mouse intestinal organoids a significant impact of IFN-λ signaling in intestinal epithelial cells and neutrophils to T. gondii control in the gastrointestinal tract. Our results expand the repertoire of interferons that contribute to the control of T. gondii and may lead to novel therapeutic approaches against this world-wide zoonotic pathogen.
Collapse
Affiliation(s)
- Mateo Murillo-León
- Institute of Virology, Medical Center University of Freiburg, 79104 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Aura M. Bastidas-Quintero
- Institute of Virology, Medical Center University of Freiburg, 79104 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Niklas S. Endres
- Institute of Virology, Medical Center University of Freiburg, 79104 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
- Current address:Institute of Virology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Daniel Schnepf
- Institute of Virology, Medical Center University of Freiburg, 79104 Freiburg, Germany
- Current address: Immunoregulation Laboratory, The Francis Crick Institute, London, UK
| | | | - Annette Ohnemus
- Institute of Virology, Medical Center University of Freiburg, 79104 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Gregory A. Taylor
- Departments of Medicine; Molecular Genetics and Microbiology; and Immunology; and Center for the Study of Aging and Human Development, Duke University Medical Center, NC 27710 Durham, North Carolina, United States of America
- Geriatric Research, Education, and Clinical Center, Durham VA Health Care System, NC 27705 Durham, North Carolina, United States of America
| | - Martin Schwemmle
- Institute of Virology, Medical Center University of Freiburg, 79104 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Peter Staeheli
- Institute of Virology, Medical Center University of Freiburg, 79104 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Tobias Steinfeldt
- Institute of Virology, Medical Center University of Freiburg, 79104 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| |
Collapse
|
7
|
Steinhäuser S, Silva P, Lenk L, Beder T, Hartmann A, Hänzelmann S, Fransecky L, Neumann M, Bastian L, Lipinski S, Richter K, Bultmann M, Hübner E, Xia S, Röllig C, Vogiatzi F, Schewe DM, Yumiceba V, Schultz K, Spielmann M, Baldus CD. Isocitrate dehydrogenase 1 mutation drives leukemogenesis by PDGFRA activation due to insulator disruption in acute myeloid leukemia (AML). Leukemia 2023; 37:134-142. [PMID: 36411356 PMCID: PMC9883162 DOI: 10.1038/s41375-022-01751-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 10/24/2022] [Accepted: 10/28/2022] [Indexed: 11/22/2022]
Abstract
Acute myeloid leukemia (AML) is characterized by complex molecular alterations and driver mutations. Elderly patients show increased frequencies of IDH mutations with high chemoresistance and relapse rates despite recent therapeutic advances. Besides being associated with global promoter hypermethylation, IDH1 mutation facilitated changes in 3D DNA-conformation by CTCF-anchor methylation and upregulated oncogene expression in glioma, correlating with poor prognosis. Here, we investigated the role of IDH1 p.R132H mutation in altering 3D DNA-architecture and subsequent oncogene activation in AML. Using public RNA-Seq data, we identified upregulation of tyrosine kinase PDGFRA in IDH1-mutant patients, correlating with poor prognosis. DNA methylation analysis identified CpG hypermethylation within a CTCF-anchor upstream of PDGFRA in IDH1-mutant patients. Increased PDGFRA expression, PDGFRA-CTCF methylation and decreased CTCF binding were confirmed in AML CRISPR cells with heterozygous IDH1 p.R132H mutation and upon exogenous 2-HG treatment. IDH1-mutant cells showed higher sensitivity to tyrosine kinase inhibitor dasatinib, which was supported by reduced blast count in a patient with refractory IDH1-mutant AML after dasatinib treatment. Our data illustrate that IDH1 p.R132H mutation leads to CTCF hypermethylation, disrupting DNA-looping and insulation of PDGFRA, resulting in PDGFRA upregulation in IDH1-mutant AML. Treatment with dasatinib may offer a novel treatment strategy for IDH1-mutant AML.
Collapse
Affiliation(s)
- Sophie Steinhäuser
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Patricia Silva
- Department of Hematology and Oncology, Charité University Hospital, Berlin, Germany
| | - Lennart Lenk
- Department of Pediatrics I, ALL-BFM Study Group, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Thomas Beder
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Alina Hartmann
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Sonja Hänzelmann
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Lars Fransecky
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Martin Neumann
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Lorenz Bastian
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Simone Lipinski
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
- University Cancer Center Schleswig-Holstein (UCCSH), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Kathrin Richter
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Miriam Bultmann
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Emely Hübner
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Shuli Xia
- Kennedy Krieger Institute, Baltimore, MD, USA
- School of Medicine, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Christoph Röllig
- Department of Internal Medicine I, University Hospital Carl-Gustav-Carus, Dresden, Germany
| | - Fotini Vogiatzi
- Department of Pediatrics I, ALL-BFM Study Group, University Hospital Schleswig-Holstein, Kiel, Germany
| | | | - Veronica Yumiceba
- Institute for Human Genetics, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Kristin Schultz
- Institute for Human Genetics, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Malte Spielmann
- Institute for Human Genetics, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Claudia Dorothea Baldus
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany.
- University Cancer Center Schleswig-Holstein (UCCSH), University Hospital Schleswig-Holstein, Kiel, Germany.
| |
Collapse
|
8
|
Sailor KA, Agoranos G, López-Manzaneda S, Tada S, Gillet-Legrand B, Guerinot C, Masson JB, Vestergaard CL, Bonner M, Gagnidze K, Veres G, Lledo PM, Cartier N. Hematopoietic stem cell transplantation chemotherapy causes microglia senescence and peripheral macrophage engraftment in the brain. Nat Med 2022; 28:517-527. [PMID: 35190726 DOI: 10.1038/s41591-022-01691-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 01/10/2022] [Indexed: 02/07/2023]
|
9
|
Hossain FMA, Park SO, Kim HJ, Eo JC, Choi JY, Tanveer M, Uyangaa E, Kim K, Eo SK. Indoleamine 2,3-Dioxygenase in Hematopoietic Stem Cell-Derived Cells Suppresses Rhinovirus-Induced Neutrophilic Airway Inflammation by Regulating Th1- and Th17-Type Responses. Immune Netw 2021; 21:e26. [PMID: 34522439 PMCID: PMC8410990 DOI: 10.4110/in.2021.21.e26] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/31/2021] [Accepted: 08/05/2021] [Indexed: 12/11/2022] Open
Abstract
Asthma exacerbations are a major cause of intractable morbidity, increases in health care costs, and a greater progressive loss of lung function. Asthma exacerbations are most commonly triggered by respiratory viral infections, particularly with human rhinovirus (hRV). Respiratory viral infections are believed to affect the expression of indoleamine 2,3-dioxygenase (IDO), a limiting enzyme in tryptophan catabolism, which is presumed to alter asthmatic airway inflammation. Here, we explored the detailed role of IDO in the progression of asthma exacerbations using a mouse model for asthma exacerbation caused by hRV infection. Our results reveal that IDO is required to prevent neutrophilic inflammation in the course of asthma exacerbation caused by an hRV infection, as corroborated by markedly enhanced Th17- and Th1-type neutrophilia in the airways of IDO-deficient mice. This neutrophilia was closely associated with disrupted expression of tight junctions and enhanced expression of inflammasome-related molecules and mucin-inducing genes. In addition, IDO ablation enhanced allergen-specific Th17- and Th1-biased CD4+ T-cell responses following hRV infection. The role of IDO in attenuating Th17- and Th1-type neutrophilic airway inflammation became more apparent in chronic asthma exacerbations after repeated allergen exposures and hRV infections. Furthermore, IDO enzymatic induction in leukocytes derived from the hematopoietic stem cell (HSC) lineage appeared to play a dominant role in attenuating Th17- and Th1-type neutrophilic inflammation in the airway following hRV infection. Therefore, IDO activity in HSC-derived leukocytes is required to regulate Th17- and Th1-type neutrophilic inflammation in the airway during asthma exacerbations caused by hRV infections.
Collapse
Affiliation(s)
- Ferdaus Mohd Altaf Hossain
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan 54596, Korea.,Faculty of Veterinary, Animal and Biomedical Sciences, Sylhet Agricultural University, Sylhet 3100, Bangladesh
| | - Seong Ok Park
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan 54596, Korea
| | - Hyo Jin Kim
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan 54596, Korea
| | - Jun Cheol Eo
- Division of Biotechnology, College of Environmental & Biosource Science, Jeonbuk National University, Iksan 54596, Korea
| | - Jin Young Choi
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan 54596, Korea
| | - Maryum Tanveer
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan 54596, Korea
| | - Erdenebelig Uyangaa
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan 54596, Korea
| | - Koanhoi Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Seong Kug Eo
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan 54596, Korea
| |
Collapse
|
10
|
Orsi M, Palmai-Pallag M, Yakoub Y, Ibouraadaten S, De Beukelaer M, Bouzin C, Bearzatto B, Ambroise J, Gala JL, Brusa D, Lison D, Huaux F. Monocytic Ontogeny of Regenerated Macrophages Characterizes the Mesotheliomagenic Responses to Carbon Nanotubes. Front Immunol 2021; 12:666107. [PMID: 34194430 PMCID: PMC8236701 DOI: 10.3389/fimmu.2021.666107] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/28/2021] [Indexed: 12/13/2022] Open
Abstract
Macrophages are not only derived from circulating blood monocytes or embryonic precursors but also expand by proliferation. The origin determines macrophage fate and functions in steady state and pathological conditions. Macrophages predominantly infiltrate fibre-induced mesothelioma tumors and contribute to cancer development. Here, we revealed their ontogeny by comparing the response to needle-like mesotheliomagenic carbon nanotubes (CNT-7) with tangled-like non-mesotheliomagenic CNT-T. In a rat peritoneal cavity model of mesothelioma, both CNT induced a rapid macrophage disappearance reaction (MDR) of MHCIIlow resident macrophages generating an empty niche available for macrophage repopulation. Macrophage depletion after mesotheliomagenic CNT-7 was followed by a substantial inflammatory reaction, and macrophage replenishment completed after 7 days. Thirty days after non-mesotheliomagenic CNT-T, macrophage repopulation was still incomplete and accompanied by a limited inflammatory reaction. Cell depletion experiments, flow cytometry and RNA-seq analysis demonstrated that, after mesotheliomagenic CNT-7 exposure, resident macrophages were mainly replaced by an influx of monocytes, which differentiated locally into MHCIIhigh inflammatory macrophages. In contrast, the low inflammatory response induced by CNT-T was associated by the accumulation of self-renewing MHCIIlow macrophages that initially derive from monocytes. In conclusion, the mesotheliomagenic response to CNT specifically relies on macrophage niche recolonization by monocyte-derived inflammatory macrophages. In contrast, the apparent homeostasis after non-mesotheliomagenic CNT treatment involves a macrophage regeneration by proliferation. Macrophage depletion and repopulation are thus decisive events characterizing the carcinogenic activity of particles and fibres.
Collapse
Affiliation(s)
- Micaela Orsi
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Mihaly Palmai-Pallag
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Yousof Yakoub
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Saloua Ibouraadaten
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Michèle De Beukelaer
- Imaging Platform, Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Caroline Bouzin
- Imaging Platform, Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Bertrand Bearzatto
- Center for Applied Molecular Technologies, Institute of Experimental and Clinical Research (IREC), Cliniques Universitaires Saint-Luc and Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Jérôme Ambroise
- Center for Applied Molecular Technologies, Institute of Experimental and Clinical Research (IREC), Cliniques Universitaires Saint-Luc and Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Jean-Luc Gala
- Center for Applied Molecular Technologies, Institute of Experimental and Clinical Research (IREC), Cliniques Universitaires Saint-Luc and Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Davide Brusa
- Flow Cytometry Platform, Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Dominique Lison
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - François Huaux
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
11
|
Ochi K, Morita M, Wilkinson AC, Iwama A, Yamazaki S. Non-conditioned bone marrow chimeric mouse generation using culture-based enrichment of hematopoietic stem and progenitor cells. Nat Commun 2021; 12:3568. [PMID: 34117255 PMCID: PMC8195984 DOI: 10.1038/s41467-021-23763-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 05/08/2021] [Indexed: 12/13/2022] Open
Abstract
Bone marrow (BM) chimeric mice are a valuable tool in the field of immunology, with the genetic manipulation of donor cells widely used to study gene function under physiological and pathological settings. To date, however, BM chimera protocols require myeloablative conditioning of recipient mice, which dramatically alters steady-state hematopoiesis. Additionally, most protocols use fluorescence-activated cell sorting (FACS) of hematopoietic stem/progenitor cells (HSPCs) for ex vivo genetic manipulation. Here, we describe our development of cell culture techniques for the enrichment of functional HSPCs from mouse BM without the use of FACS purification. Furthermore, the large number of HSPCs derived from these cultures generate BM chimeric mice without irradiation. These HSPC cultures can also be genetically manipulated by viral transduction, to allow for doxycycline-inducible transgene expression in donor-derived immune cells within non-conditioned immunocompetent recipients. This technique is therefore expected to overcome current limitations in mouse transplantation models.
Collapse
Affiliation(s)
- Kiyosumi Ochi
- Division of Stem Cell Biology, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Maiko Morita
- Division of Stem Cell Biology, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Adam C Wilkinson
- MRC Molecular Hematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Atsushi Iwama
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Satoshi Yamazaki
- Division of Stem Cell Biology, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
- Laboratory of Stem Cell Therapy, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan.
| |
Collapse
|
12
|
El-Serafi A, He R, Zheng W, Benkossou F, Oerther S, Zhao Y, Mellgren K, Gustafsson B, Heilmann C, Kanerva J, Lotfi K, Toporski J, Sundin M, Höglund M, Mattsson J, El-Serafi I, Hassan M. Vitamin D levels and busulphan kinetics in patients undergoing hematopoietic stem cell transplantation, a multicenter study. Bone Marrow Transplant 2021; 56:807-817. [PMID: 33087877 DOI: 10.1038/s41409-020-01091-y] [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: 05/14/2020] [Revised: 09/24/2020] [Accepted: 10/08/2020] [Indexed: 11/09/2022]
Abstract
Vitamin D (Vit-D), an essential nutrient, interacts with different drugs including chemotherapeutic agents like busulphan, an alkylating agent used for conditioning prior to stem cell transplantation. The correlation between Vit-D plasma levels and busulphan clearance was investigated in an uncontrolled prospective study in patients and mice. Plasma 25(OH)D levels were measured and busulphan pharmacokinetics calculated in 81 patients. Adults received oral busulphan (n = 34) while children received busulphan orally (n = 19) or intravenously (n = 28). Patients received no Vit-D supplementation. To confirm our findings, pharmacokinetics after a single dose of busulphan (oral or intravenous) were evaluated in two groups of mice (n = 60) receiving high or standard-level Vit-D supplementation. Both busulphan clearance (P < 0.0001) and 25(OH)D levels (P = 0.0004) were significantly higher in adults compared to children. A significant negative correlation (P = 0.041) was found between busulphan clearance and 25(OH)D levels in children treated orally. No such correlation was observed in adults or in children receiving intravenous busulphan. In addition, no significant effect of Vit-D levels on busulphan pharmacokinetics in mice regardless of the administration route. In conclusion, 25(OH)D can affect oral busulphan pharmacokinetics in children and its level should be considered when personalizing oral busulphan treatment. Further studies are warranted to confirm the underlying mechanisms.
Collapse
Affiliation(s)
- Ahmed El-Serafi
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Rui He
- Experimental Cancer Medicine, Division of Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Clinical Research Center and Center of Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Wenyi Zheng
- Experimental Cancer Medicine, Division of Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Clinical Research Center and Center of Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Fadwa Benkossou
- Experimental Cancer Medicine, Division of Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Clinical Research Center and Center of Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Sandra Oerther
- Experimental Cancer Medicine, Division of Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Pre-clinical Laboratory, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Ying Zhao
- Experimental Cancer Medicine, Division of Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Clinical Research Center and Center of Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Karin Mellgren
- Department of Pediatric Oncology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Britt Gustafsson
- Department of Clinical Science, Intervention and Technology, CLINTEC, Karolinska Institutet, Stockholm, Sweden
| | - Carsten Heilmann
- Pediatric Clinic, Rigshospitalet, National University Hospital, Copenhagen, Denmark
| | - Jukka Kanerva
- HUS Helsinki University Hospital and University of Helsinki, New Children's Hospital, Division of Hematology-Oncology and Stem Cell Transplantation, Helsinki, Finland
| | - Kourosh Lotfi
- Clinical Pharmacology, Division of Drug Research, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
- Department of Hematology, Linköping University Hospital, Linköping, Sweden
| | - Jacek Toporski
- Department of Pediatrics, Skåne University Hospital, Lund, Sweden
| | - Mikael Sundin
- Division of Pediatrics, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
- Section of Pediatric Hematology, Immunology and HCT, Astrid Lindgren Children's Hospital, Stockholm, Sweden
| | - Martin Höglund
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Jonas Mattsson
- Gloria and Seymour Epstein Chair in Cell Therapy and Transplantation, University of Toronto, Toronto, Canada
- Princess Margaret Cancer Centre, Toronto, ON, Canada
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Ibrahim El-Serafi
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Biochemistry, Faculty of Medicine, Port-Said University, Port-Said, Egypt
| | - Moustapha Hassan
- Experimental Cancer Medicine, Division of Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.
- Clinical Research Center and Center of Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden.
| |
Collapse
|
13
|
Li Q, Lan X, Han X, Durham F, Wan J, Weiland A, Koehler RC, Wang J. Microglia-derived interleukin-10 accelerates post-intracerebral hemorrhage hematoma clearance by regulating CD36. Brain Behav Immun 2021; 94:437-457. [PMID: 33588074 PMCID: PMC8058329 DOI: 10.1016/j.bbi.2021.02.001] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 12/13/2020] [Accepted: 02/06/2021] [Indexed: 12/19/2022] Open
Abstract
Hematoma size after intracerebral hemorrhage (ICH) significantly affects patient outcome. However, our knowledge of endogenous mechanisms that underlie hematoma clearance and the potential role of the anti-inflammatory cytokine interleukin-10 (IL-10) is limited. Using organotypic hippocampal slice cultures and a collagenase-induced ICH mouse model, we investigated the role of microglial IL-10 in phagocytosis ex vivo and hematoma clearance in vivo. In slice culture, exposure to hemoglobin induced IL-10 expression in microglia and enhanced phagocytosis that depended on IL-10-regulated expression of CD36. Following ICH, IL-10-deficient mice had more severe neuroinflammation, brain edema, iron deposition, and neurologic deficits associated with delayed hematoma clearance. Intranasal administration of recombinant IL-10 accelerated hematoma clearance and improved neurologic function. Additionally, IL-10-deficient mice had weakened in vivo phagocytic ability owing to decreased expression of microglial CD36. Moreover, loss of IL-10 significantly increased monocyte-derived macrophage infiltration and enhanced brain inflammation in vivo. These results indicate that IL-10 regulates microglial phagocytosis and monocyte-derived macrophage infiltration after ICH and that CD36 is a key phagocytosis effector regulated by IL-10. Leveraging the innate immune response to ICH by augmenting IL-10 signaling may provide a useful strategy for accelerating hematoma clearance and improving neurologic outcome in clinical translation studies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Raymond C. Koehler
- Corresponding author at: Department of
Anesthesiology and Critical Care Medicine, The Johns Hopkins University School
of Medicine, 600 North Wolfe Street Blalock 1404, Baltimore, MD 21287, USA,
(R.C. Koehler)
| | | |
Collapse
|
14
|
Hohsfield LA, Najafi AR, Ghorbanian Y, Soni N, Hingco EE, Kim SJ, Jue AD, Swarup V, Inlay MA, Green KN. Effects of long-term and brain-wide colonization of peripheral bone marrow-derived myeloid cells in the CNS. J Neuroinflammation 2020; 17:279. [PMID: 32951604 PMCID: PMC7504855 DOI: 10.1186/s12974-020-01931-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/17/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Microglia, the primary resident myeloid cells of the brain, play critical roles in immune defense by maintaining tissue homeostasis and responding to injury or disease. However, microglial activation and dysfunction has been implicated in a number of central nervous system (CNS) disorders, thus developing tools to manipulate and replace these myeloid cells in the CNS is of therapeutic interest. METHODS Using whole body irradiation, bone marrow transplant, and colony-stimulating factor 1 receptor inhibition, we achieve long-term and brain-wide (~ 80%) engraftment and colonization of peripheral bone marrow-derived myeloid cells (i.e., monocytes) in the brain parenchyma and evaluated the long-term effects of their colonization in the CNS. RESULTS Here, we identify a monocyte signature that includes an upregulation in Ccr1, Ms4a6b, Ms4a6c, Ms4a7, Apobec1, Lyz2, Mrc1, Tmem221, Tlr8, Lilrb4a, Msr1, Nnt, and Wdfy1 and a downregulation of Siglech, Slc2a5, and Ccl21a/b. We demonstrate that irradiation and long-term (~ 6 months) engraftment of the CNS by monocytes induces brain region-dependent alterations in transcription profiles, astrocytes, neuronal structures, including synaptic components, and cognition. Although our results show that microglial replacement with peripherally derived myeloid cells is feasible and that irradiation-induced changes can be reversed by the replacement of microglia with monocytes in the hippocampus, we also observe that brain-wide engraftment of peripheral myeloid cells (relying on irradiation) can result in cognitive and synaptic deficits. CONCLUSIONS These findings provide insight into better understanding the role and complexity of myeloid cells in the brain, including their regulation of other CNS cells and functional outcomes.
Collapse
Affiliation(s)
- Lindsay A Hohsfield
- Department of Neurobiology and Behavior, University of California, 3208 Biological Sciences III, Irvine, CA, 92697-4545, USA
| | - Allison R Najafi
- Department of Neurobiology and Behavior, University of California, 3208 Biological Sciences III, Irvine, CA, 92697-4545, USA
| | - Yasamine Ghorbanian
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, 92697, USA
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, 92697, USA
| | - Neelakshi Soni
- Department of Neurobiology and Behavior, University of California, 3208 Biological Sciences III, Irvine, CA, 92697-4545, USA
| | - Edna E Hingco
- Department of Neurobiology and Behavior, University of California, 3208 Biological Sciences III, Irvine, CA, 92697-4545, USA
| | - Sung Jin Kim
- Department of Neurobiology and Behavior, University of California, 3208 Biological Sciences III, Irvine, CA, 92697-4545, USA
| | - Ayer Darling Jue
- Department of Neurobiology and Behavior, University of California, 3208 Biological Sciences III, Irvine, CA, 92697-4545, USA
| | - Vivek Swarup
- Department of Neurobiology and Behavior, University of California, 3208 Biological Sciences III, Irvine, CA, 92697-4545, USA
| | - Mathew A Inlay
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, 92697, USA
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, 92697, USA
| | - Kim N Green
- Department of Neurobiology and Behavior, University of California, 3208 Biological Sciences III, Irvine, CA, 92697-4545, USA.
| |
Collapse
|
15
|
Contribution of a GATA4-Expressing Hematopoietic Progenitor Lineage to the Adult Mouse Endothelium. Cells 2020; 9:cells9051257. [PMID: 32438714 PMCID: PMC7290801 DOI: 10.3390/cells9051257] [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: 03/30/2020] [Revised: 05/17/2020] [Accepted: 05/18/2020] [Indexed: 11/17/2022] Open
Abstract
Different sources have been claimed for the embryonic origin of the coronary endothelium. Recently, the potential of circulating cells as progenitors of the cardiac endothelium has also been suggested. In a previous study we have shown that circulating progenitors are recruited by the embryonic endocardium and incorporated into the coronary vessels. These progenitors derive from a mesodermal lineage characterized by the expression of Gata4 under control of the enhancer G2. Herein, we aim to trace this specific lineage throughout postnatal stages. We have found that more than 50% of the adult cardiac endothelium derives from the G2-GATA4 lineage. This percentage increases from embryos to adults probably due to differential proliferation and postnatal recruitment of circulating endothelial progenitors. In fact, injection of fetal liver or placental cells in the blood stream of neonates leads to incorporation of G2-GATA4 lineage cells to the coronary endothelium. On the other hand, labeling of the hematopoietic lineage by the stage E7.5 also resulted in positive coronary endothelial cells from both, embryos and adults. Our results suggest that early hematopoietic progenitors recruited by the embryonic ventricular endocardium can become the predominant source of definitive endothelium during the vascularization of the heart.
Collapse
|
16
|
Ferreira FM, Palle P, Vom Berg J, Prajwal P, Laman JD, Buch T. Bone marrow chimeras-a vital tool in basic and translational research. J Mol Med (Berl) 2019; 97:889-896. [PMID: 31028417 DOI: 10.1007/s00109-019-01783-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 03/26/2019] [Accepted: 04/01/2019] [Indexed: 12/16/2022]
Abstract
Bone marrow chimeras are used routinely in immunology research as well as in other fields of biology. Here, we provide a concise state-of-the-art review about the types of chimerisms that can be achieved and the type of information that each model generates. We include separate sections for caveats and future developments. We provide examples from the literature in which different types of chimerism were employed to answer specific questions. While simple bone marrow chimeras allow to dissect the role of genes in distinct cell populations such as the hematopoietic cells versus non-hematopoietic cells, mixed bone marrow chimeras can provide detailed information about hematopoietic cell types and the intrinsic and extrinsic roles of individual genes. The advantages and caveats of bone marrow chimerism for the study of microglia are addressed, as well as alternatives to irradiation that minimize blood-brain-barrier disruption. Elementary principles are introduced and their potential is exemplified through summarizing recent studies.
Collapse
Affiliation(s)
- Filipa M Ferreira
- Institute of Laboratory Animal Science, University of Zurich, Zurich, Switzerland
| | | | - Johannes Vom Berg
- Institute of Laboratory Animal Science, University of Zurich, Zurich, Switzerland
| | - Prajwal Prajwal
- Institute of Laboratory Animal Science, University of Zurich, Zurich, Switzerland
| | - Jon D Laman
- Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands
| | - Thorsten Buch
- Institute of Laboratory Animal Science, University of Zurich, Zurich, Switzerland. .,Institute of Laboratory Animal Science, University of Zurich, Wagistrasse 12, 8952, Schlieren, Switzerland.
| |
Collapse
|
17
|
Regoli M, Man A, Gicheva N, Dumont A, Ivory K, Pacini A, Morucci G, Branca JJV, Lucattelli M, Santosuosso U, Narbad A, Gulisano M, Bertelli E, Nicoletti C. Morphological and Functional Characterization of IL-12Rβ2 Chain on Intestinal Epithelial Cells: Implications for Local and Systemic Immunoregulation. Front Immunol 2018; 9:1177. [PMID: 29896198 PMCID: PMC5986899 DOI: 10.3389/fimmu.2018.01177] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 05/11/2018] [Indexed: 01/01/2023] Open
Abstract
Interaction between intestinal epithelial cells (IECs) and the underlying immune systems is critical for maintaining intestinal immune homeostasis and mounting appropriate immune responses. We have previously showed that the T helper type 1 (TH1) cytokine IL-12 plays a key role in the delicate immunological balance in the gut and the lack of appropriate levels of IL-12 had important consequences for health and disease, particularly with regard to food allergy. Here, we sought to understand the role of IL-12 in the regulation of lymphoepithelial cross talk and how this interaction affects immune responses locally and systemically. Using a combination of microscopy and flow cytometry techniques we observed that freshly isolated IECs expressed an incomplete, yet functional IL-12 receptor (IL-12R) formed solely by the IL-12Rβ2 chain that albeit the lack of the complementary IL-12β1 chain responded to ex vivo challenge with IL-12. Furthermore, the expression of IL-12Rβ2 on IECs is strategically located at the interface between epithelial and immune cells of the lamina propria and using in vitro coculture models and primary intestinal organoids we showed that immune-derived signals were required for the expression of IL-12Rβ2 on IECs. The biological relevance of the IEC-associated IL-12Rβ2 was assessed in vivo in a mouse model of food allergy characterized by allergy-associated diminished intestinal levels of IL-12 and in chimeric mice that lack the IL-12Rβ2 chain on IECs. These experimental models enabled us to show that the antiallergic properties of orally delivered recombinant Lactococcus lactis secreting bioactive IL-12 (rLc-IL12) were reduced in mice lacking the IL-12β2 chain on IECs. Finally, we observed that the oral delivery of IL-12 was accompanied by the downregulation of the production of the IEC-derived proallergic cytokine thymic stromal lymphopoietin (TSLP). However, further analysis of intestinal levels of TSLP in IL-12Rβ2−/− mice suggested that this event was not directly linked to the IEC-associated IL-12Rβ2 chain. We interpreted these data as showing that IEC-associated IL12Rβ2 is a component of the cytokine network operating at the interface between the intestinal epithelium and immune system that plays a role in immune regulation.
Collapse
Affiliation(s)
- Mari Regoli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Angela Man
- Gut Health Programme, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Nadhezda Gicheva
- Gut Health Programme, Quadram Institute Bioscience, Norwich, United Kingdom
| | | | - Kamal Ivory
- Gut Health Programme, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Alessandra Pacini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Gabriele Morucci
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Jacopo J V Branca
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Monica Lucattelli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Ugo Santosuosso
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Arjan Narbad
- Gut Health Programme, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Massimo Gulisano
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Eugenio Bertelli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Claudio Nicoletti
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
18
|
Peake K, Manning J, Lewis CA, Tran K, Rossi F, Krieger C. Bone Marrow-Derived Cell Accumulation in the Spinal Cord Is Independent of Peripheral Mobilization in a Mouse Model of Amyotrophic Lateral Sclerosis. Front Neurol 2017; 8:75. [PMID: 28337172 PMCID: PMC5340765 DOI: 10.3389/fneur.2017.00075] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 02/20/2017] [Indexed: 12/14/2022] Open
Abstract
Bone marrow-derived cells (BMDCs) are capable of migrating across the blood–brain barrier (BBB) and accumulating in the central nervous system (CNS) when transplanted into recipients conditioned with whole-body irradiation or chemotherapy. We used the chemotherapeutic agents busulfan and treosulfan to condition recipient mice for transplantation with bone marrow (BM) cells isolated from donor mice ubiquitously expressing green fluorescent protein. We attempted to increase the accumulation of BMDCs in the CNS by mobilization of BMDCs using either, or both, granulocyte colony-stimulating factor (GCSF) or plerixafor (AMD3100). We also used several concentrations of busulfan. We hypothesized that higher concentrations of busulfan and BMDC mobilization would increase numbers of GFP+ cells in the CNS. The doses of busulfan employed (60–125 mg/kg) all resulted in high levels of sustained chimerism (>85% 1 year post-transplant) in both the blood and BM of wild-type (WT) mice and an amyotrophic lateral sclerosis (ALS) mouse model. Moreover, cells accumulated within the CNS in a dose-, time-, and disease-dependent manner. Conditioning with the hydrophilic busulfan analog treosulfan, which is unable to cross the BBB efficiently, also resulted in a high degree of BM chimerism. However, few GFP+ BMDCs were found within the CNS of WT or ALS mice of treosulfan-conditioned mice. Mobilization of BMDCs into the circulation using GCSF and/or AMD3100 did not lead to increased accumulation of GFP+ BMDCs within the CNS of WT or ALS mice. Weekly analysis of BMDC accumulation revealed that BMDCs accumulated more rapidly and to a greater extent in the CNS of ALS mice conditioned with a high dose (125 mg/kg) of busulfan compared to a lower dose (80 mg/kg). The number of GFP+ BMDCs in the CNS labeling with the proliferation marker Ki67 increased in parallel with BMDC accumulation within the CNS. Our results indicate that establishment of high levels of blood and BM chimerism alone is not sufficient to induce BMDC accumulation within the CNS and that CNS conditioning is a crucial requirement for BMDC accumulation to occur. Moreover, it appears that proliferation of BMDCs that infiltrate the CNS is partly responsible for cell accumulation in busulfan-conditioned ALS mice.
Collapse
Affiliation(s)
- Kyle Peake
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University , Burnaby, BC , Canada
| | - John Manning
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University , Burnaby, BC , Canada
| | - Coral-Ann Lewis
- The Biomedical Research Centre, University of British Columbia , Vancouver, BC , Canada
| | - Kevin Tran
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University , Burnaby, BC , Canada
| | - Fabio Rossi
- The Biomedical Research Centre, University of British Columbia , Vancouver, BC , Canada
| | - Charles Krieger
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada; Division of Neurology, Department of Medicine, Neuromuscular Disease Unit, VHHSC, Vancouver, BC, Canada
| |
Collapse
|
19
|
Das A, Ghatak S, Sinha M, Chaffee S, Ahmed NS, Parinandi NL, Wohleb ES, Sheridan JF, Sen CK, Roy S. Correction of MFG-E8 Resolves Inflammation and Promotes Cutaneous Wound Healing in Diabetes. THE JOURNAL OF IMMUNOLOGY 2016; 196:5089-100. [PMID: 27194784 DOI: 10.4049/jimmunol.1502270] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 04/18/2016] [Indexed: 12/15/2022]
Abstract
Milk fat globule epidermal growth factor-factor 8 (MFG-E8) is a peripheral glycoprotein that acts as a bridging molecule between the macrophage and apoptotic cells, thus executing a pivotal role in the scavenging of apoptotic cells from affected tissue. We have previously reported that apoptotic cell clearance activity or efferocytosis is compromised in diabetic wound macrophages. In this work, we test the hypothesis that MFG-E8 helps resolve inflammation, supports angiogenesis, and accelerates wound closure. MFG-E8(-/-) mice displayed impaired efferocytosis associated with exaggerated inflammatory response, poor angiogenesis, and wound closure. Wound macrophage-derived MFG-E8 was recognized as a critical driver of wound angiogenesis. Transplantation of MFG-E8(-/-) bone marrow to MFG-E8(+/+) mice resulted in impaired wound closure and compromised wound vascularization. In contrast, MFG-E8(-/-) mice that received wild-type bone marrow showed improved wound closure and improved wound vascularization. Hyperglycemia and exposure to advanced glycated end products inactivated MFG-E8, recognizing a key mechanism that complicates diabetic wound healing. Diabetic db/db mice suffered from impaired efferocytosis accompanied with persistent inflammation and slow wound closure. Topical recombinant MFG-E8 induced resolution of wound inflammation, improvements in angiogenesis, and acceleration of closure, upholding the potential of MFG-E8-directed therapeutics in diabetic wound care.
Collapse
Affiliation(s)
- Amitava Das
- Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210; Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Comprehensive Wound Center, Center for Regenerative Medicine and Cell Based Therapies, Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Subhadip Ghatak
- Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210; Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Comprehensive Wound Center, Center for Regenerative Medicine and Cell Based Therapies, Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Mithun Sinha
- Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210; Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Comprehensive Wound Center, Center for Regenerative Medicine and Cell Based Therapies, Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Scott Chaffee
- Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210; Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Comprehensive Wound Center, Center for Regenerative Medicine and Cell Based Therapies, Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Noha S Ahmed
- Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210; Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Comprehensive Wound Center, Center for Regenerative Medicine and Cell Based Therapies, Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Narasimham L Parinandi
- Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Department of Internal Medicine, Ohio State University Wexner Medical Center, Columbus, OH 43210; and
| | - Eric S Wohleb
- Division of Biosciences, The Ohio State University, Columbus, OH 43210
| | - John F Sheridan
- Division of Biosciences, The Ohio State University, Columbus, OH 43210
| | - Chandan K Sen
- Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210; Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Comprehensive Wound Center, Center for Regenerative Medicine and Cell Based Therapies, Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Sashwati Roy
- Department of Surgery, Ohio State University Wexner Medical Center, Columbus, OH 43210; Davis Heart and Lung Research Institute, Ohio State University Wexner Medical Center, Columbus, OH 43210; Comprehensive Wound Center, Center for Regenerative Medicine and Cell Based Therapies, Ohio State University Wexner Medical Center, Columbus, OH 43210;
| |
Collapse
|