101
|
Zhou H, Zeng Z, Koentgen F, Khan M, Mombaerts P. The testicular soma of Tsc22d3 knockout mice supports spermatogenesis and germline transmission from spermatogonial stem cell lines upon transplantation. Genesis 2019; 57:e23295. [PMID: 31001916 PMCID: PMC6617806 DOI: 10.1002/dvg.23295] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 02/19/2019] [Accepted: 03/22/2019] [Indexed: 12/16/2022]
Abstract
Spermatogonial stem cells (SSCs) are adult stem cells that are slowly cycling and self-renewing. The pool of SSCs generates very large numbers of male gametes throughout the life of the individual. SSCs can be cultured in vitro for long periods of time, and established SSC lines can be manipulated genetically. Upon transplantation into the testes of infertile mice, long-term cultured mouse SSCs can differentiate into fertile spermatozoa, which can give rise to live offspring. Here, we show that the testicular soma of mice with a conditional knockout (conKO) in the X-linked gene Tsc22d3 supports spermatogenesis and germline transmission from cultured mouse SSCs upon transplantation. Infertile males were produced by crossing homozygous Tsc22d3 floxed females with homozygous ROSA26-Cre males. We obtained 96 live offspring from six long-term cultured SSC lines with the aid of intracytoplasmic sperm injection. We advocate the further optimization of Tsc22d3-conKO males as recipients for testis transplantation of SSC lines.
Collapse
Affiliation(s)
- Hai Zhou
- Max Planck Research Unit for Neurogenetics, Frankfurt, Germany
| | - Zhen Zeng
- Max Planck Research Unit for Neurogenetics, Frankfurt, Germany
| | | | - Mona Khan
- Max Planck Research Unit for Neurogenetics, Frankfurt, Germany
| | - Peter Mombaerts
- Max Planck Research Unit for Neurogenetics, Frankfurt, Germany
| |
Collapse
|
102
|
Frisch BJ, Hoffman CM, Latchney SE, LaMere MW, Myers J, Ashton J, Li AJ, Saunders J, Palis J, Perkins AS, McCabe A, Smith JN, McGrath KE, Rivera-Escalera F, McDavid A, Liesveld JL, Korshunov VA, Elliott MR, MacNamara KC, Becker MW, Calvi LM. Aged marrow macrophages expand platelet-biased hematopoietic stem cells via Interleukin1B. JCI Insight 2019; 5:124213. [PMID: 30998506 DOI: 10.1172/jci.insight.124213] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The bone marrow microenvironment (BMME) contributes to the regulation of hematopoietic stem cell (HSC) function, though its role in age-associated lineage skewing is poorly understood. Here we show that dysfunction of aged marrow macrophages (Mφs) directs HSC platelet-bias. Mφs from the marrow of aged mice and humans exhibited an activated phenotype, with increased expression of inflammatory signals. Aged marrow Mφs also displayed decreased phagocytic function. Senescent neutrophils, typically cleared by marrow Mφs, were markedly increased in aged mice, consistent with functional defects in Mφ phagocytosis and efferocytosis. In aged mice, Interleukin 1B (IL1B) was elevated in the bone marrow and caspase 1 activity, which can process pro-IL1B, was increased in marrow Mφs and neutrophils. Mechanistically, IL1B signaling was necessary and sufficient to induce a platelet bias in HSCs. In young mice, depletion of phagocytic cell populations or loss of the efferocytic receptor Axl expanded platelet-biased HSCs. Our data support a model wherein increased inflammatory signals and decreased phagocytic function of aged marrow Mφs induce the acquisition of platelet bias in aged HSCs. This work highlights the instructive role of Mφs and IL1B in the age-associated lineage-skewing of HSCs, and reveals the therapeutic potential of their manipulation as antigeronic targets.
Collapse
Affiliation(s)
| | - Corey M Hoffman
- Department of Medicine.,James P. Wilmot Cancer Institute, and.,Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | | | - Mark W LaMere
- Department of Medicine.,James P. Wilmot Cancer Institute, and
| | - Jason Myers
- James P. Wilmot Cancer Institute, and.,UR Genomics Research Center, Rochester, New York, USA
| | - John Ashton
- James P. Wilmot Cancer Institute, and.,UR Genomics Research Center, Rochester, New York, USA
| | - Allison J Li
- Department of Medicine.,James P. Wilmot Cancer Institute, and
| | - Jerry Saunders
- Center for Pediatric Biomedical Research and.,Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - James Palis
- James P. Wilmot Cancer Institute, and.,Center for Pediatric Biomedical Research and
| | - Archibald S Perkins
- James P. Wilmot Cancer Institute, and.,Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Amanda McCabe
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Julianne Np Smith
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | | | | | - Andrew McDavid
- Department of Biostatistics and Computational Biology, and
| | - Jane L Liesveld
- Department of Medicine.,James P. Wilmot Cancer Institute, and
| | - Vyacheslav A Korshunov
- Department of Medicine.,Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, New York, USA
| | | | - Katherine C MacNamara
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | | | - Laura M Calvi
- Department of Medicine.,James P. Wilmot Cancer Institute, and
| |
Collapse
|
103
|
Boyer SW, Rajendiran S, Beaudin AE, Smith-Berdan S, Muthuswamy PK, Perez-Cunningham J, Martin EW, Cheung C, Tsang H, Landon M, Forsberg EC. Clonal and Quantitative In Vivo Assessment of Hematopoietic Stem Cell Differentiation Reveals Strong Erythroid Potential of Multipotent Cells. Stem Cell Reports 2019; 12:801-815. [PMID: 30905737 PMCID: PMC6450035 DOI: 10.1016/j.stemcr.2019.02.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/16/2019] [Accepted: 02/18/2019] [Indexed: 01/22/2023] Open
Abstract
Hematopoiesis is arguably one of the best understood stem cell systems; however, significant challenges remain to reach a consensus understanding of the lineage potential, heterogeneity, and relationships of hematopoietic stem and progenitor cell populations. To gain new insights, we performed quantitative analyses of mature cell production from hematopoietic stem cells (HSCs) and multiple hematopoietic progenitor populations. Assessment of the absolute numbers of mature cell types produced by each progenitor cell revealed a striking erythroid dominance of all myeloid-competent progenitors assessed, accompanied by strong platelet reconstitution. All populations with myeloid potential also produced robust numbers of red blood cells and platelets in vivo. Clonal analysis by single-cell transplantation and by spleen colony assays revealed that a significant fraction of HSCs and multipotent progenitors have multilineage potential at the single-cell level. These new insights prompt an erythroid-focused model of hematopoietic differentiation. RBCs are the predominant cell type produced by multipotent hematopoietic progenitors All cell types with myeloid potential also produced RBCs and platelets in vivo Single HSCs and MPPF cells are capable of multilineage hematopoietic reconstitution Erythroid cell production emerges as a default hematopoietic fate
Collapse
Affiliation(s)
- Scott W Boyer
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Smrithi Rajendiran
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Anna E Beaudin
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Stephanie Smith-Berdan
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Praveen K Muthuswamy
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Jessica Perez-Cunningham
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Eric W Martin
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Christa Cheung
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Herman Tsang
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Mark Landon
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - E Camilla Forsberg
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA.
| |
Collapse
|
104
|
A miR-150/TET3 pathway regulates the generation of mouse and human non-classical monocyte subset. Nat Commun 2018; 9:5455. [PMID: 30575719 PMCID: PMC6303340 DOI: 10.1038/s41467-018-07801-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 11/19/2018] [Indexed: 12/30/2022] Open
Abstract
Non-classical monocyte subsets may derive from classical monocyte differentiation and the proportion of each subset is tightly controlled. Deregulation of this repartition is observed in diverse human diseases, including chronic myelomonocytic leukemia (CMML) in which non-classical monocyte numbers are significantly decreased relative to healthy controls. Here, we identify a down-regulation of hsa-miR-150 through methylation of a lineage-specific promoter in CMML monocytes. Mir150 knock-out mice demonstrate a cell-autonomous defect in non-classical monocytes. Our pulldown experiments point to Ten-Eleven-Translocation-3 (TET3) mRNA as a hsa-miR-150 target in classical human monocytes. We show that Tet3 knockout mice generate an increased number of non-classical monocytes. Our results identify the miR-150/TET3 axis as being involved in the generation of non-classical monocytes.
Collapse
|
105
|
Lainšček D, Kadunc L, Keber MM, Bratkovič IH, Romih R, Jerala R. Delivery of an Artificial Transcription Regulator dCas9-VPR by Extracellular Vesicles for Therapeutic Gene Activation. ACS Synth Biol 2018; 7:2715-2725. [PMID: 30513193 DOI: 10.1021/acssynbio.8b00192] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The CRISPR/Cas system has been developed as a potent tool for genome engineering and transcription regulation. However, the efficiency of the delivery of the system into cells, particularly for therapeutic in vivo applications, remains a major bottleneck. Extracellular vesicles (EVs), released by eukaryotic cells, can mediate the transfer of various molecules, including nucleic acids and proteins. We show the packaging and delivery of the CRISPR/Cas system via EVs to the target cells, combining the advantages of both technological platforms. A genome editing with designed extracellular vesicles (GEDEX) system generated by the producer cells can transfer the designed transcriptional regulator dCas9-VPR complexed with appropriate targeting gRNAs enabling activation of gene transcription. We show functional delivery in mammalian cells as well in the animals. The therapeutic efficiency of in vivo delivery of dCas9-VPR/sgRNA GEDEX is demonstrated in a mouse model of liver damage counteracted by upregulation of the endogenous hepatocyte growth factor, demonstrating the potential for therapeutic applications.
Collapse
Affiliation(s)
- Duško Lainšček
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana 1000, Slovenia
| | - Lucija Kadunc
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana 1000, Slovenia
- Graduate School of Biomedicine, University of Ljubljana, Ljubljana 1000, Slovenia
| | - Mateja Manček Keber
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana 1000, Slovenia
- EN-FIST Centre of Excellence, Trg Osvobodilne fronte 13, Ljubljana 1000, Slovenia
| | - Iva Hafner Bratkovič
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana 1000, Slovenia
| | - Rok Romih
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, Ljubljana 1000, Slovenia
| | - Roman Jerala
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana 1000, Slovenia
- EN-FIST Centre of Excellence, Trg Osvobodilne fronte 13, Ljubljana 1000, Slovenia
| |
Collapse
|
106
|
Kotov DI, Pengo T, Mitchell JS, Gastinger MJ, Jenkins MK. Chrysalis: A New Method for High-Throughput Histo-Cytometry Analysis of Images and Movies. THE JOURNAL OF IMMUNOLOGY 2018; 202:300-308. [PMID: 30510065 DOI: 10.4049/jimmunol.1801202] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 11/02/2018] [Indexed: 12/17/2022]
Abstract
Advances in imaging have led to the development of powerful multispectral, quantitative imaging techniques, like histo-cytometry. The utility of this approach is limited, however, by the need for time consuming manual image analysis. We therefore developed the software Chrysalis and a group of Imaris Xtensions to automate this process. The resulting automation allowed for high-throughput histo-cytometry analysis of three-dimensional confocal microscopy and two-photon time-lapse images of T cell-dendritic cell interactions in mouse spleens. It was also applied to epi-fluorescence images to quantify T cell localization within splenic tissue by using a "signal absorption" strategy that avoids computationally intensive distance measurements. In summary, this image processing and analysis software makes histo-cytometry more useful for immunology applications by automating image analysis.
Collapse
Affiliation(s)
- Dmitri I Kotov
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455; .,Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Thomas Pengo
- University of Minnesota Informatics Institute, University of Minnesota Twin Cities, Minneapolis, MN 55455
| | - Jason S Mitchell
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455.,University Imaging Centers, University of Minnesota, Minneapolis, MN 55455.,Department of Medicine, University of Minnesota, Minneapolis, MN 55455; and
| | | | - Marc K Jenkins
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455.,Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
107
|
Impaired hematopoiesis and leukemia development in mice with a conditional knock-in allele of a mutant splicing factor gene U2af1. Proc Natl Acad Sci U S A 2018; 115:E10437-E10446. [PMID: 30322915 PMCID: PMC6217397 DOI: 10.1073/pnas.1812669115] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Somatic mutations in some splicing factor genes are frequently found in myelodysplastic syndromes (MDS) and MDS-related acute myeloid leukemia (AML), blood cancers with few effective treatment options. However, the pathophysiological effects of these mutations remain poorly characterized. Here, we report the establishment of mouse models to study a common splicing factor mutation, U2AF1(S34F). Production of the mutant protein in the murine hematopoietic compartment disrupts hematopoiesis in ways resembling human MDS. We further identified deletion of the Runx1 gene and other known oncogenic mutations as changes that might collaborate with U2af1(S34F) to give rise to frank AML in mice. However, the U2af1(S34F) mutation was absent in two of the three AML cases, raising the possibility that this mutant protein plays a dispensable role in tumor maintenance. Mutations affecting the spliceosomal protein U2AF1 are commonly found in myelodysplastic syndromes (MDS) and secondary acute myeloid leukemia (sAML). We have generated mice that carry Cre-dependent knock-in alleles of U2af1(S34F), the murine version of the most common mutant allele of U2AF1 encountered in human cancers. Cre-mediated recombination in murine hematopoietic lineages caused changes in RNA splicing, as well as multilineage cytopenia, macrocytic anemia, decreased hematopoietic stem and progenitor cells, low-grade dysplasias, and impaired transplantability, but without lifespan shortening or leukemia development. In an attempt to identify U2af1(S34F)-cooperating changes that promote leukemogenesis, we combined U2af1(S34F) with Runx1 deficiency in mice and further treated the mice with a mutagen, N-ethyl-N-nitrosourea (ENU). Overall, 3 of 16 ENU-treated compound transgenic mice developed AML. However, AML did not arise in mice with other genotypes or without ENU treatment. Sequencing DNA from the three AMLs revealed somatic mutations homologous to those considered to be drivers of human AML, including predicted loss- or gain-of-function mutations in Tet2, Gata2, Idh1, and Ikzf1. However, the engineered U2af1(S34F) missense mutation reverted to WT in two of the three AML cases, implying that U2af1(S34F) is dispensable, or even selected against, once leukemia is established.
Collapse
|
108
|
Boulais PE, Mizoguchi T, Zimmerman S, Nakahara F, Vivié J, Mar JC, van Oudenaarden A, Frenette PS. The Majority of CD45 - Ter119 - CD31 - Bone Marrow Cell Fraction Is of Hematopoietic Origin and Contains Erythroid and Lymphoid Progenitors. Immunity 2018; 49:627-639.e6. [PMID: 30314756 DOI: 10.1016/j.immuni.2018.08.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 04/13/2018] [Accepted: 08/24/2018] [Indexed: 01/01/2023]
Abstract
The non-hematopoietic cell fraction of the bone marrow (BM) is classically identified as CD45- Ter119- CD31- (herein referred to as triple-negative cells or TNCs). Although TNCs are believed to contain heterogeneous stromal cell populations, they remain poorly defined. Here we showed that the vast majority of TNCs (∼85%) have a hematopoietic rather than mesenchymal origin. Single cell RNA-sequencing revealed erythroid and lymphoid progenitor signatures among CD51- TNCs. Ly6D+ CD44+ CD51- TNCs phenotypically and functionally resembled CD45+ pro-B lymphoid cells, whereas Ly6D- CD44+ CD51- TNCs were enriched in previously unappreciated stromal-dependent erythroid progenitors hierarchically situated between preCFU-E and proerythroblasts. Upon adoptive transfer, CD44+ CD51- TNCs contributed to repopulate the B-lymphoid and erythroid compartments. CD44+ CD51- TNCs also expanded during phenylhydrazine-induced acute hemolysis or in a model of sickle cell anemia. These findings thus uncover physiologically relevant new classes of stromal-associated functional CD45- hematopoietic progenitors.
Collapse
Affiliation(s)
- Philip E Boulais
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, 10461, USA; The Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Toshihide Mizoguchi
- Institute for Oral Science, Matsumoto Dental University, Nagano, 399-0781 Japan
| | - Samuel Zimmerman
- The Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York, 10461, USA; Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Fumio Nakahara
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, 10461, USA; The Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Judith Vivié
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, Cancer Genomics Netherlands, 3584 CT Utrecht, the Netherlands
| | - Jessica C Mar
- The Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York, 10461, USA; Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane QLD 4072, Australia
| | - Alexander van Oudenaarden
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, Cancer Genomics Netherlands, 3584 CT Utrecht, the Netherlands
| | - Paul S Frenette
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, 10461, USA; Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, 10461, USA; The Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York, 10461, USA.
| |
Collapse
|
109
|
Roediger B, Lee Q, Tikoo S, Cobbin JCA, Henderson JM, Jormakka M, O'Rourke MB, Padula MP, Pinello N, Henry M, Wynne M, Santagostino SF, Brayton CF, Rasmussen L, Lisowski L, Tay SS, Harris DC, Bertram JF, Dowling JP, Bertolino P, Lai JH, Wu W, Bachovchin WW, Wong JJL, Gorrell MD, Shaban B, Holmes EC, Jolly CJ, Monette S, Weninger W. An Atypical Parvovirus Drives Chronic Tubulointerstitial Nephropathy and Kidney Fibrosis. Cell 2018; 175:530-543.e24. [PMID: 30220458 PMCID: PMC6800251 DOI: 10.1016/j.cell.2018.08.013] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 07/23/2018] [Accepted: 08/07/2018] [Indexed: 11/19/2022]
Abstract
The occurrence of a spontaneous nephropathy with intranuclear inclusions in laboratory mice has puzzled pathologists for over 4 decades, because its etiology remains elusive. The condition is more severe in immunodeficient animals, suggesting an infectious cause. Using metagenomics, we identify the causative agent as an atypical virus, termed "mouse kidney parvovirus" (MKPV), belonging to a divergent genus of Parvoviridae. MKPV was identified in animal facilities in Australia and North America, is transmitted via a fecal-oral or urinary-oral route, and is controlled by the adaptive immune system. Detailed analysis of the clinical course and histopathological features demonstrated a stepwise progression of pathology ranging from sporadic tubular inclusions to tubular degeneration and interstitial fibrosis and culminating in renal failure. In summary, we identify a widely distributed pathogen in laboratory mice and establish MKPV-induced nephropathy as a new tool for elucidating mechanisms of tubulointerstitial fibrosis that shares molecular features with chronic kidney disease in humans.
Collapse
Affiliation(s)
- Ben Roediger
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia.
| | - Quintin Lee
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Shweta Tikoo
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Joanna C A Cobbin
- Marie Bashir Institute for Infectious Diseases and Biosecurity, School of Life and Environmental Sciences and Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - James M Henderson
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Mika Jormakka
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Matthew B O'Rourke
- Mass Spectrometry Core Facility, University of Sydney, Sydney, NSW 2006, Australia; Proteomics Core Facility, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Matthew P Padula
- Proteomics Core Facility, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Natalia Pinello
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Marisa Henry
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia; Laboratory Animal Services, University of Sydney, Sydney, NSW 2006, Australia
| | - Maria Wynne
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia; Laboratory Animal Services, University of Sydney, Sydney, NSW 2006, Australia
| | - Sara F Santagostino
- Laboratory of Comparative Pathology, Center of Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center, The Rockefeller University, Weill Cornell Medicine, New York, NY 10065, USA
| | - Cory F Brayton
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | - Leszek Lisowski
- Children's Medical Research Institute, University of Sydney, Sydney, NSW 2006, Australia; Military Institute of Hygiene and Epidemiology, Biological Threats Identification and Countermeasure Centre, Puławy 24-100, Poland
| | - Szun S Tay
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
| | - David C Harris
- Centre for Transplantation and Renal Research, Westmead Institute for Medical Research, University of Sydney, NSW 2006, Australia
| | - John F Bertram
- Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC 3800, Australia
| | - John P Dowling
- Department of Anatomical Pathology, Monash Medical Centre, Clayton, VIC 3168, Australia
| | - Patrick Bertolino
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Jack H Lai
- Sackler School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Wengen Wu
- Sackler School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA
| | - William W Bachovchin
- Sackler School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Justin J-L Wong
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Mark D Gorrell
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Babak Shaban
- Australian Genomics Research Facility, Parkville, VIC 3000, Australia; Melbourne Integrative Genomics, University of Melbourne, Parkville, VIC 3010, Australia
| | - Edward C Holmes
- Marie Bashir Institute for Infectious Diseases and Biosecurity, School of Life and Environmental Sciences and Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Christopher J Jolly
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Sébastien Monette
- Laboratory of Comparative Pathology, Center of Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center, The Rockefeller University, Weill Cornell Medicine, New York, NY 10065, USA
| | - Wolfgang Weninger
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia; Discipline of Dermatology, Faculty of Medicine and Health, University of Sydney, NSW 2006, Australia; Department of Dermatology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia; Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria.
| |
Collapse
|
110
|
A Metabolism-Based Quorum Sensing Mechanism Contributes to Termination of Inflammatory Responses. Immunity 2018; 49:654-665.e5. [DOI: 10.1016/j.immuni.2018.07.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 04/26/2018] [Accepted: 07/24/2018] [Indexed: 12/22/2022]
|
111
|
Shen Y, Cohen JL, Nicoloro SM, Kelly M, Yenilmez B, Henriques F, Tsagkaraki E, Edwards YJK, Hu X, Friedline RH, Kim JK, Czech MP. CRISPR-delivery particles targeting nuclear receptor-interacting protein 1 ( Nrip1) in adipose cells to enhance energy expenditure. J Biol Chem 2018; 293:17291-17305. [PMID: 30190322 DOI: 10.1074/jbc.ra118.004554] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 08/22/2018] [Indexed: 12/26/2022] Open
Abstract
RNA-guided, engineered nucleases derived from the prokaryotic adaptive immune system CRISPR-Cas represent a powerful platform for gene deletion and editing. When used as a therapeutic approach, direct delivery of Cas9 protein and single-guide RNA (sgRNA) could circumvent the safety issues associated with plasmid delivery and therefore represents an attractive tool for precision genome engineering. Gene deletion or editing in adipose tissue to enhance its energy expenditure, fatty acid oxidation, and secretion of bioactive factors through a "browning" process presents a potential therapeutic strategy to alleviate metabolic disease. Here, we developed "CRISPR-delivery particles," denoted CriPs, composed of nano-size complexes of Cas9 protein and sgRNA that are coated with an amphipathic peptide called Endo-Porter that mediates entry into cells. Efficient CRISPR-Cas9-mediated gene deletion of ectopically expressed GFP by CriPs was achieved in multiple cell types, including a macrophage cell line, primary macrophages, and primary pre-adipocytes. Significant GFP loss was also observed in peritoneal exudate cells with minimum systemic toxicity in GFP-expressing mice following intraperitoneal injection of CriPs containing Gfp-targeting sgRNA. Furthermore, disruption of a nuclear co-repressor of catabolism, the Nrip1 gene, in white adipocytes by CriPs enhanced adipocyte browning with a marked increase of uncoupling protein 1 (UCP1) expression. Of note, the CriP-mediated Nrip1 deletion did not produce detectable off-target effects. We conclude that CriPs offer an effective Cas9 and sgRNA delivery system for ablating targeted gene products in cultured cells and in vivo, providing a potential therapeutic strategy for metabolic disease.
Collapse
Affiliation(s)
| | | | | | - Mark Kelly
- From the Program in Molecular Medicine and
| | | | | | - Emmanouela Tsagkaraki
- From the Program in Molecular Medicine and.,the Molecular Basis of Human Disease Graduate Program, School of Sciences, Faculty of Medicine, University of Crete, P.O. Box 2208, Heraklion, Crete 71003, Greece
| | | | - Xiaodi Hu
- the Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605 and
| | - Randall H Friedline
- the Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605 and
| | - Jason K Kim
- From the Program in Molecular Medicine and.,the Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605 and
| | | |
Collapse
|
112
|
Aging alters the immunological response to ischemic stroke. Acta Neuropathol 2018; 136:89-110. [PMID: 29752550 PMCID: PMC6015099 DOI: 10.1007/s00401-018-1859-2] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 04/21/2018] [Accepted: 04/30/2018] [Indexed: 12/15/2022]
Abstract
The peripheral immune system plays a critical role in aging and in the response to brain injury. Emerging data suggest inflammatory responses are exacerbated in older animals following ischemic stroke; however, our understanding of these age-related changes is poor. In this work, we demonstrate marked differences in the composition of circulating and infiltrating leukocytes recruited to the ischemic brain of old male mice after stroke compared to young male mice. Blood neutrophilia and neutrophil invasion into the brain were increased in aged animals. Relative to infiltrating monocyte populations, brain-invading neutrophils had reduced phagocytic potential, and produced higher levels of reactive oxygen species and extracellular matrix-degrading enzymes (i.e., MMP-9), which were further exacerbated with age. Hemorrhagic transformation was more pronounced in aged versus young mice relative to infarct size. High numbers of myeloperoxidase-positive neutrophils were found in postmortem human brain samples of old (> 71 years) acute ischemic stroke subjects compared to non-ischemic controls. Many of these neutrophils were found in the brain parenchyma. A large proportion of these neutrophils expressed MMP-9 and positively correlated with hemorrhage and hyperemia. MMP-9 expression and hemorrhagic transformation after stroke increased with age. These changes in the myeloid response to stroke with age led us to hypothesize that the bone marrow response to stroke is altered with age, which could be important for the development of effective therapies targeting the immune response. We generated heterochronic bone marrow chimeras as a tool to determine the contribution of peripheral immune senescence to age- and stroke-induced inflammation. Old hosts that received young bone marrow (i.e., Young → Old) had attenuation of age-related reductions in bFGF and VEGF and showed improved locomotor activity and gait dynamics compared to isochronic (Old → Old) controls. Microglia in young heterochronic mice (Old → Young) developed a senescent-like phenotype. After stroke, aged animals reconstituted with young marrow had reduced behavioral deficits compared to isochronic controls, and had significantly fewer brain-infiltrating neutrophils. Increased rates of hemorrhagic transformation were seen in young mice reconstituted with aged bone marrow. This work suggests that age alters the immunological response to stroke, and that this can be reversed by manipulation of the peripheral immune cells in the bone marrow.
Collapse
|
113
|
Bolden JE, Lucas EC, Zhou G, O'Sullivan JA, de Graaf CA, McKenzie MD, Di Rago L, Baldwin TM, Shortt J, Alexander WS, Bochner BS, Ritchie ME, Hilton DJ, Fairfax KA. Identification of a Siglec-F+ granulocyte-macrophage progenitor. J Leukoc Biol 2018; 104:123-133. [PMID: 29645346 PMCID: PMC6320667 DOI: 10.1002/jlb.1ma1217-475r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 02/14/2018] [Accepted: 02/16/2018] [Indexed: 01/09/2023] Open
Abstract
In recent years multi-parameter flow cytometry has enabled identification of cells at major stages in myeloid development; from pluripotent hematopoietic stem cells, through populations with increasingly limited developmental potential (common myeloid progenitors and granulocyte-macrophage progenitors), to terminally differentiated mature cells. Myeloid progenitors are heterogeneous, and the surface markers that define transition states from progenitors to mature cells are poorly characterized. Siglec-F is a surface glycoprotein frequently used in combination with IL-5 receptor alpha (IL5Rα) for the identification of murine eosinophils. Here, we describe a CD11b+ Siglec-F+ IL5Rα- myeloid population in the bone marrow of C57BL/6 mice. The CD11b+ Siglec-F+ IL5Rα- cells are retained in eosinophil deficient PHIL mice, and are not expanded upon overexpression of IL-5, indicating that they are upstream or independent of the eosinophil lineage. We show these cells to have GMP-like developmental potential in vitro and in vivo, and to be transcriptionally distinct from the classically described GMP population. The CD11b+ Siglec-F+ IL5Rα- population expands in the bone marrow of Myb mutant mice, which is potentially due to negative transcriptional regulation of Siglec-F by Myb. Lastly, we show that the role of Siglec-F may be, at least in part, to regulate GMP viability.
Collapse
Affiliation(s)
- Jessica E Bolden
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Erin C Lucas
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Geyu Zhou
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Jeremy A O'Sullivan
- Division of Allergy and Immunology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Carolyn A de Graaf
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Mark D McKenzie
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Ladina Di Rago
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Tracey M Baldwin
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Jake Shortt
- School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia
| | - Warren S Alexander
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Bruce S Bochner
- Division of Allergy and Immunology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Matthew E Ritchie
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Douglas J Hilton
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Kirsten A Fairfax
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
114
|
Biomimetic post-capillary venule expansions for leukocyte adhesion studies. Sci Rep 2018; 8:9328. [PMID: 29921896 PMCID: PMC6008471 DOI: 10.1038/s41598-018-27566-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 06/05/2018] [Indexed: 02/02/2023] Open
Abstract
Leukocyte adhesion and extravasation are maximal near the transition from capillary to post-capillary venule, and are strongly influenced by a confluence of scale-dependent physical effects. Mimicking the scale of physiological vessels using in vitro microfluidic systems allows the capture of these effects on leukocyte adhesion assays, but imposes practical limits on reproducibility and reliable quantification. Here we present a microfluidic platform that provides multiple (54-512) technical replicates within a 15-minute sample collection time, coupled with an automated computer vision analysis pipeline that captures leukocyte adhesion probabilities as a function of shear and extensional stresses. We report that in post-capillary channels of physiological scale, efficient leukocyte adhesion requires erythrocytes forcing leukocytes against the wall, a phenomenon that is promoted by the transitional flow in post-capillary venule expansions and dependent on the adhesion molecule ICAM-1.
Collapse
|
115
|
Moalli F, Ficht X, Germann P, Vladymyrov M, Stolp B, de Vries I, Lyck R, Balmer J, Fiocchi A, Kreutzfeldt M, Merkler D, Iannacone M, Ariga A, Stoffel MH, Sharpe J, Bähler M, Sixt M, Diz-Muñoz A, Stein JV. The Rho regulator Myosin IXb enables nonlymphoid tissue seeding of protective CD8 + T cells. J Exp Med 2018; 215:1869-1890. [PMID: 29875261 PMCID: PMC6028505 DOI: 10.1084/jem.20170896] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 12/28/2017] [Accepted: 05/11/2018] [Indexed: 12/27/2022] Open
Abstract
Moalli et al. combine in vitro CD8+ T cell motility analysis with intravital imaging of mouse tissues to identify the actomyosin regulator Myo9b as a central player for nonlymphoid tissue infiltration during adaptive immune responses by facilitating crossing of tissue barriers. T cells are actively scanning pMHC-presenting cells in lymphoid organs and nonlymphoid tissues (NLTs) with divergent topologies and confinement. How the T cell actomyosin cytoskeleton facilitates this task in distinct environments is incompletely understood. Here, we show that lack of Myosin IXb (Myo9b), a negative regulator of the small GTPase Rho, led to increased Rho-GTP levels and cell surface stiffness in primary T cells. Nonetheless, intravital imaging revealed robust motility of Myo9b−/− CD8+ T cells in lymphoid tissue and similar expansion and differentiation during immune responses. In contrast, accumulation of Myo9b−/− CD8+ T cells in NLTs was strongly impaired. Specifically, Myo9b was required for T cell crossing of basement membranes, such as those which are present between dermis and epidermis. As consequence, Myo9b−/− CD8+ T cells showed impaired control of skin infections. In sum, we show that Myo9b is critical for the CD8+ T cell adaptation from lymphoid to NLT surveillance and the establishment of protective tissue–resident T cell populations.
Collapse
Affiliation(s)
- Federica Moalli
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Xenia Ficht
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Philipp Germann
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain.,European Molecular Biology Laboratory, Barcelona, Spain
| | - Mykhailo Vladymyrov
- Albert Einstein Center for Fundamental Physics, Laboratory for High Energy Physics (LHEP), University of Bern, Bern, Switzerland
| | - Bettina Stolp
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Ingrid de Vries
- Institute for Science and Technology Austria, Klosterneuburg, Austria
| | - Ruth Lyck
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Jasmin Balmer
- Department of Clinical Research and Veterinary Public Health, University of Bern, Bern, Switzerland
| | - Amleto Fiocchi
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Mario Kreutzfeldt
- Department of Pathology and Immunology, Division of Clinical Pathology, University and University Hospitals of Geneva, Geneva, Switzerland
| | - Doron Merkler
- Department of Pathology and Immunology, Division of Clinical Pathology, University and University Hospitals of Geneva, Geneva, Switzerland
| | - Matteo Iannacone
- Division of Immunology, Transplantation and Infectious Diseases and Experimental Imaging Center, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Milan, Italy
| | - Akitaka Ariga
- Albert Einstein Center for Fundamental Physics, Laboratory for High Energy Physics (LHEP), University of Bern, Bern, Switzerland
| | - Michael H Stoffel
- Department of Clinical Research and Veterinary Public Health, University of Bern, Bern, Switzerland
| | - James Sharpe
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain.,European Molecular Biology Laboratory, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Martin Bähler
- Institut für Molekulare Zellbiologie, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Michael Sixt
- Institute for Science and Technology Austria, Klosterneuburg, Austria
| | - Alba Diz-Muñoz
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Jens V Stein
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| |
Collapse
|
116
|
Hons M, Kopf A, Hauschild R, Leithner A, Gaertner F, Abe J, Renkawitz J, Stein JV, Sixt M. Chemokines and integrins independently tune actin flow and substrate friction during intranodal migration of T cells. Nat Immunol 2018; 19:606-616. [PMID: 29777221 DOI: 10.1038/s41590-018-0109-z] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Accepted: 04/11/2018] [Indexed: 01/13/2023]
Abstract
Although much is known about the physiological framework of T cell motility, and numerous rate-limiting molecules have been identified through loss-of-function approaches, an integrated functional concept of T cell motility is lacking. Here, we used in vivo precision morphometry together with analysis of cytoskeletal dynamics in vitro to deconstruct the basic mechanisms of T cell migration within lymphatic organs. We show that the contributions of the integrin LFA-1 and the chemokine receptor CCR7 are complementary rather than positioned in a linear pathway, as they are during leukocyte extravasation from the blood vasculature. Our data demonstrate that CCR7 controls cortical actin flows, whereas integrins mediate substrate friction that is sufficient to drive locomotion in the absence of considerable surface adhesions and plasma membrane flux.
Collapse
Affiliation(s)
- Miroslav Hons
- Institute of Science and Technology Austria, Klosterneuburg, Austria.
- Theodor Kocher Institute, University of Bern, Bern, Switzerland.
| | - Aglaja Kopf
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Robert Hauschild
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | | | - Florian Gaertner
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Jun Abe
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Jörg Renkawitz
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Jens V Stein
- Theodor Kocher Institute, University of Bern, Bern, Switzerland.
| | - Michael Sixt
- Institute of Science and Technology Austria, Klosterneuburg, Austria.
| |
Collapse
|
117
|
Clonal expansion and myeloid leukemia progression modeled by multiplex gene editing of murine hematopoietic progenitor cells. Exp Hematol 2018; 64:33-44.e5. [PMID: 29751067 DOI: 10.1016/j.exphem.2018.04.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 04/24/2018] [Accepted: 04/25/2018] [Indexed: 01/22/2023]
Abstract
Recent advances in next-generation sequencing have identified novel mutations and revealed complex genetic architectures in human hematological malignancies. Moving forward, new methods to quickly generate animal models that recapitulate the complex genetics of human hematological disorders are needed to transform the genetic information to new therapies. Here, we used a ribonucleoprotein-based CRISPR/Cas9 system to model human clonal hematopoiesis of indeterminate potential and acute myeloid leukemia (AML). We edited multiple genes recurrently mutated in hematological disorders, including those encoding epigenetic regulators, transcriptional regulators, and signaling components in murine hematopoietic stem/progenitor cells. Tracking the clonal dynamics by sequencing the indels induced by CRISPR/Cas9 revealed clonal expansion in some recipient mice that progressed to AML initiated by leukemia-initiating cells. Our results establish that the CRISPR/Cas9-mediated multiplex mutagenesis can be used to engineer a variety of murine models of hematological malignancies with complex genetic architectures seen in human disease.
Collapse
|
118
|
Cabinian A, Sinsimer D, Tang M, Jang Y, Choi B, Laouar Y, Laouar A. Gut symbiotic microbes imprint intestinal immune cells with the innate receptor SLAMF4 which contributes to gut immune protection against enteric pathogens. Gut 2018; 67:847-859. [PMID: 28341747 PMCID: PMC5890651 DOI: 10.1136/gutjnl-2016-313214] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 02/27/2017] [Accepted: 03/04/2017] [Indexed: 12/25/2022]
Abstract
BACKGROUND Interactions between host immune cells and gut microbiota are crucial for the integrity and function of the intestine. How these interactions regulate immune cell responses in the intestine remains a major gap in the field. AIM We have identified the signalling lymphocyte activation molecule family member 4 (SLAMF4) as an immunomodulator of the intestinal immunity. The aim is to determine how SLAMF4 is acquired in the gut and what its contribution to intestinal immunity is. METHODS Expression of SLAMF4 was assessed in mice and humans. The mechanism of induction was studied using GFPtg bone marrow chimaera mice, lymphotoxin α and TNLG8A-deficient mice, as well as gnotobiotic mice. Role in immune protection was revealed using oral infection with Listeria monocytogenes and Cytobacter rodentium. RESULTS SLAMF4 is a selective marker of intestinal immune cells of mice and humans. SLAMF4 induction occurs directly in the intestinal mucosa without the involvement of the gut-associated lymphoid tissue. Gut bacterial products, particularly those of gut anaerobes, and gut-resident antigen-presenting cell (APC) TNLG8A are key contributors of SLAMF4 induction in the intestine. Importantly, lack of SLAMF4 expression leads the increased susceptibility of mice to infection by oral pathogens culminating in their premature death. CONCLUSIONS SLAMF4 is a marker of intestinal immune cells which contributes to the protection against enteric pathogens and whose expression is dependent on the presence of the gut microbiota. This discovery provides a possible mechanism for answering the long-standing question of how the intertwining of the host and gut microbial biology regulates immune cell responses in the gut.
Collapse
Affiliation(s)
- Allison Cabinian
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA
| | - Daniel Sinsimer
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA
| | - May Tang
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA
| | - Youngsoon Jang
- Department of Microbiology and Immunology, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Bongkum Choi
- Department of Microbiology and Immunology, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Yasmina Laouar
- Department of Microbiology and Immunology, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Amale Laouar
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA
| |
Collapse
|
119
|
De Niz M, Meibalan E, Mejia P, Ma S, Brancucci NMB, Agop-Nersesian C, Mandt R, Ngotho P, Hughes KR, Waters AP, Huttenhower C, Mitchell JR, Martinelli R, Frischknecht F, Seydel KB, Taylor T, Milner D, Heussler VT, Marti M. Plasmodium gametocytes display homing and vascular transmigration in the host bone marrow. SCIENCE ADVANCES 2018; 4:eaat3775. [PMID: 29806032 PMCID: PMC5966192 DOI: 10.1126/sciadv.aat3775] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 04/12/2018] [Indexed: 05/13/2023]
Abstract
Transmission of Plasmodium parasites to the mosquito requires the formation and development of gametocytes. Studies in infected humans have shown that only the most mature forms of Plasmodium falciparum gametocytes are present in circulation, whereas immature forms accumulate in the hematopoietic environment of the bone marrow. We used the rodent model Plasmodium berghei to study gametocyte behavior through time under physiological conditions. Intravital microscopy demonstrated preferential homing of early gametocyte forms across the intact vascular barrier of the bone marrow and the spleen early during infection and subsequent development in the extravascular environment. During the acute phase of infection, we observed vascular leakage resulting in further parasite accumulation in this environment. Mature gametocytes showed high deformability and were found entering and exiting the intact vascular barrier. We suggest that extravascular gametocyte localization and mobility are essential for gametocytogenesis and transmission of Plasmodium to the mosquito.
Collapse
Affiliation(s)
- Mariana De Niz
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA 02115, USA
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, CH-3012 Bern, Switzerland
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, G12 8TA Scotland, UK
| | - Elamaran Meibalan
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA 02115, USA
| | - Pedro Mejia
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA 02115, USA
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Siyuan Ma
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Nicolas M. B. Brancucci
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA 02115, USA
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, G12 8TA Scotland, UK
| | - Carolina Agop-Nersesian
- Department of Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, MA 02118, USA
| | - Rebecca Mandt
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA 02115, USA
| | - Priscilla Ngotho
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, G12 8TA Scotland, UK
| | - Katie R. Hughes
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, G12 8TA Scotland, UK
| | - Andrew P. Waters
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, G12 8TA Scotland, UK
| | - Curtis Huttenhower
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, 415 Main Street, Cambridge, MA 02142, USA
| | - James R. Mitchell
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Roberta Martinelli
- Beth Israel Deaconess Medical Centre, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Friedrich Frischknecht
- Parasitology Centre for Infectious Diseases, University of Heidelberg Medical School, 69120 Heidelberg, Germany
| | - Karl B. Seydel
- Blantyre Malaria Project, University of Malawi College of Medicine, Blantyre 3, Malawi
- College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Terrie Taylor
- Blantyre Malaria Project, University of Malawi College of Medicine, Blantyre 3, Malawi
- College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Danny Milner
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA 02115, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
- Department of Pathology, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115, USA
| | - Volker T. Heussler
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, CH-3012 Bern, Switzerland
| | - Matthias Marti
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA 02115, USA
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, G12 8TA Scotland, UK
| |
Collapse
|
120
|
Faltusová K, Szikszai K, Molík M, Linhartová J, Páral P, Šefc L, Savvulidi F, Nečas E. Stem Cell Defect in Ubiquitin-Green Fluorescent Protein Mice Facilitates Engraftment of Lymphoid-Primed Hematopoietic Stem Cells. Stem Cells 2018; 36:1237-1248. [DOI: 10.1002/stem.2828] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 03/02/2018] [Accepted: 03/06/2018] [Indexed: 01/31/2023]
Affiliation(s)
- Kateřina Faltusová
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University
| | - Katarína Szikszai
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University
| | - Martin Molík
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University
| | - Jana Linhartová
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University
| | - Petr Páral
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University
| | - Luděk Šefc
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University
| | - Filipp Savvulidi
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University
| | - Emanuel Nečas
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University
- BIOCEV, Biotechnology and Biomedicine Center of the Academy of Sciences and Charles University in Vestec, Institute of Pathological Physiology; Czech Republic
| |
Collapse
|
121
|
Parabiosis reveals leukocyte dynamics in the kidney. J Transl Med 2018; 98:391-402. [PMID: 29251733 PMCID: PMC5839939 DOI: 10.1038/labinvest.2017.130] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Revised: 09/10/2017] [Accepted: 09/21/2017] [Indexed: 12/17/2022] Open
Abstract
The immune cellular compartment of the kidney is involved in organ development and homeostasis, as well as in many pathological conditions. Little is known about the mechanisms that drive intrarenal immune responses in the presence of renal tubular and interstitial cell death. However, it is known that tissue-resident leukocytes have the potential to have distinct roles compared with circulating cells. We used a parabiosis model in C57BL/6 CD45 congenic and green fluorescent protein transgenic mice to better understand the dynamics of immune cells in the kidney. We found F4/80Hi intrarenal macrophages exhibit minimal exchange with the peripheral circulation in two models of parabiosis, whether mice were attached for 4 or 16 weeks. Other intrarenal inflammatory cells demonstrate near total exchange with the circulating immune cell pool in healthy kidneys, indicating that innate and adaptive immune cells extensively traffic through the kidney interstitium during normal physiology. Neutrophils, dendritic cells, F4/80Low macrophages, T cells, B cells, and NK cells are renewed from the circulating immune cell pool. However, a fraction of double-negative T (CD4- CD8-) and NKT cells are long-lived or tissue resident. This study provides direct evidence of leukocyte sub-populations that are resident in the renal tissue, cells which demonstrate minimal to no exchange with the peripheral blood. In addition, the data demonstrate continual exchange of other sub-populations through uninflamed tissue.
Collapse
|
122
|
Chapple RH, Hu T, Tseng YJ, Liu L, Kitano A, Luu V, Hoegenauer KA, Iwawaki T, Li Q, Nakada D. ERα promotes murine hematopoietic regeneration through the Ire1α-mediated unfolded protein response. eLife 2018; 7:31159. [PMID: 29451493 PMCID: PMC5829925 DOI: 10.7554/elife.31159] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 02/15/2018] [Indexed: 01/08/2023] Open
Abstract
Activation of the unfolded protein response (UPR) sustains protein homeostasis (proteostasis) and plays a fundamental role in tissue maintenance and longevity of organisms. Long-range control of UPR activation has been demonstrated in invertebrates, but such mechanisms in mammals remain elusive. Here, we show that the female sex hormone estrogen regulates the UPR in hematopoietic stem cells (HSCs). Estrogen treatment increases the capacity of HSCs to regenerate the hematopoietic system upon transplantation and accelerates regeneration after irradiation. We found that estrogen signals through estrogen receptor α (ERα) expressed in hematopoietic cells to activate the protective Ire1α-Xbp1 branch of the UPR. Further, ERα-mediated activation of the Ire1α-Xbp1 pathway confers HSCs with resistance against proteotoxic stress and promotes regeneration. Our findings reveal a systemic mechanism through which HSC function is augmented for hematopoietic regeneration.
Collapse
Affiliation(s)
- Richard H Chapple
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, United States
| | - Tianyuan Hu
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, United States
| | - Yu-Jung Tseng
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, United States
| | - Lu Liu
- Division of Hematology/Oncology, Department of Medicine, University of Michigan, Ann Arbor, United States
| | - Ayumi Kitano
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, United States
| | - Victor Luu
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, United States
| | - Kevin A Hoegenauer
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, United States
| | - Takao Iwawaki
- Division of Cell Medicine, Department of Life Science, Medical Research Institute, Kanazawa Medical University, Ishikawa, Japan
| | - Qing Li
- Division of Hematology/Oncology, Department of Medicine, University of Michigan, Ann Arbor, United States
| | - Daisuke Nakada
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, United States.,Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, United States
| |
Collapse
|
123
|
Mensink M, Anstee NS, Robati M, Schenk RL, Herold MJ, Cory S, Vandenberg CJ. Anti-apoptotic A1 is not essential for lymphoma development in Eµ-Myc mice but helps sustain transplanted Eµ-Myc tumour cells. Cell Death Differ 2018; 25:797-808. [PMID: 29339775 PMCID: PMC5864240 DOI: 10.1038/s41418-017-0045-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 11/20/2017] [Accepted: 11/22/2017] [Indexed: 01/27/2023] Open
Abstract
The transcription factor c-MYC regulates a multiplicity of genes involved in cellular growth, proliferation, metabolism and DNA damage response and its overexpression is a hallmark of many tumours. Since MYC promotes apoptosis under conditions of stress, such as limited availability of nutrients or cytokines, MYC-driven cells are very much dependent on signals that inhibit cell death. Stress signals trigger apoptosis via the pathway regulated by opposing fractions of the BCL-2 protein family and previous genetic studies have shown that the development of B lymphoid tumours in Eµ-Myc mice is critically dependent on expression of pro-survival BCL-2 relatives MCL-1, BCL-W and, to a lesser extent, BCL-XL, but not BCL-2 itself, and that sustained growth of these lymphomas is dependent on MCL-1. Using recently developed mice that lack expression of all three functional pro-survival A1 genes, we show here that the kinetics of lymphoma development in Eµ-Myc mice and the competitive repopulation capacity of Eµ-Myc haemopoietic stem and progenitor cells is unaffected by the absence of A1. However, conditional loss of a single remaining functional A1 gene from transplanted A1-a−/−A1-bfl/flA1-c−/− Eµ-Myc lymphomas slowed their expansion, significantly extending the life of the transplant recipients. Thus, A1 contributes to the survival of malignant Eµ-Myc-driven B lymphoid cells. These results strengthen the case for BFL-1, the human homologue of A1, being a valid target for drug development for MYC-driven tumours.
Collapse
Affiliation(s)
- Mark Mensink
- The Walter and Eliza Hall Institute of Medical Research, Victoria, 3052, Australia.,Department of Medical Biology, University of Melbourne, Victoria, 3052, Australia.,Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Natasha S Anstee
- The Walter and Eliza Hall Institute of Medical Research, Victoria, 3052, Australia.,Department of Medical Biology, University of Melbourne, Victoria, 3052, Australia.,Deutsches Krebsforschungszentrum (DKFZ), Experimental Hematology Division, 69120, Heidelberg, Germany
| | - Mikara Robati
- The Walter and Eliza Hall Institute of Medical Research, Victoria, 3052, Australia
| | - Robyn L Schenk
- The Walter and Eliza Hall Institute of Medical Research, Victoria, 3052, Australia.,Department of Medical Biology, University of Melbourne, Victoria, 3052, Australia
| | - Marco J Herold
- The Walter and Eliza Hall Institute of Medical Research, Victoria, 3052, Australia.,Department of Medical Biology, University of Melbourne, Victoria, 3052, Australia
| | - Suzanne Cory
- The Walter and Eliza Hall Institute of Medical Research, Victoria, 3052, Australia. .,Department of Medical Biology, University of Melbourne, Victoria, 3052, Australia.
| | - Cassandra J Vandenberg
- The Walter and Eliza Hall Institute of Medical Research, Victoria, 3052, Australia.,Department of Medical Biology, University of Melbourne, Victoria, 3052, Australia
| |
Collapse
|
124
|
Abstract
Bone marrow steady-state erythropoiesis maintains erythroid homeostasis throughout life. This process constantly generates new erythrocytes to replace the senescent erythrocytes that are removed by macrophages in the spleen. In contrast, anemic or hypoxic stress induces a physiological response designed to increase oxygen delivery to the tissues. Stress erythropoiesis is a key component of this response. It is best understood in mice where it is extramedullary occurring in the adult spleen and liver and in the fetal liver during development. Stress erythropoiesis utilizes progenitor cells and signals that are distinct from bone marrow steady-state erythropoiesis. Because of that observation many genes may play a role in stress erythropoiesis despite having no effect on steady-state erythropoiesis. In this chapter, we will discuss in vivo and in vitro techniques to study stress erythropoiesis in mice and how the in vitro culture system can be extended to study human stress erythropoiesis.
Collapse
Affiliation(s)
- Laura F Bennett
- Department of Veterinary and Biomedical Sciences and Center for Molecular Immunology and Infectious Disease. Laura Bennett and Robert Paulson are Intercollege Graduate Program in Genetics. Robert Paulson and Chang Liao are Pathobiology Graduate Program, The Pennsylvania State University, 115 Henning Building, University Park, PA, 16802, USA
| | - Chang Liao
- Department of Veterinary and Biomedical Sciences and Center for Molecular Immunology and Infectious Disease. Laura Bennett and Robert Paulson are Intercollege Graduate Program in Genetics. Robert Paulson and Chang Liao are Pathobiology Graduate Program, The Pennsylvania State University, 115 Henning Building, University Park, PA, 16802, USA
| | - Robert F Paulson
- Department of Veterinary and Biomedical Sciences and Center for Molecular Immunology and Infectious Disease. Laura Bennett and Robert Paulson are Intercollege Graduate Program in Genetics. Robert Paulson and Chang Liao are Pathobiology Graduate Program, The Pennsylvania State University, 115 Henning Building, University Park, PA, 16802, USA.
| |
Collapse
|
125
|
Dietrich J, Baryawno N, Nayyar N, Valtis YK, Yang B, Ly I, Besnard A, Severe N, Gustafsson KU, Andronesi OC, Batchelor TT, Sahay A, Scadden DT. Bone marrow drives central nervous system regeneration after radiation injury. J Clin Invest 2017; 128:281-293. [PMID: 29202481 DOI: 10.1172/jci90647] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 10/24/2017] [Indexed: 01/05/2023] Open
Abstract
Nervous system injury is a frequent result of cancer therapy involving cranial irradiation, leaving patients with marked memory and other neurobehavioral disabilities. Here, we report an unanticipated link between bone marrow and brain in the setting of radiation injury. Specifically, we demonstrate that bone marrow-derived monocytes and macrophages are essential for structural and functional repair mechanisms, including regeneration of cerebral white matter and improvement in neurocognitive function. Using a granulocyte-colony stimulating factor (G-CSF) receptor knockout mouse model in combination with bone marrow cell transplantation, MRI, and neurocognitive functional assessments, we demonstrate that bone marrow-derived G-CSF-responsive cells home to the injured brain and are critical for altering neural progenitor cells and brain repair. Additionally, compared with untreated animals, animals that received G-CSF following radiation injury exhibited enhanced functional brain repair. Together, these results demonstrate that, in addition to its known role in defense and debris removal, the hematopoietic system provides critical regenerative drive to the brain that can be modulated by clinically available agents.
Collapse
Affiliation(s)
- Jorg Dietrich
- Center for Regenerative Medicine, Massachusetts General Hospital (MGH), Boston, Massachusetts, USA.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA.,Department of Neurology and Division of Neuro-Oncology, MGH, and
| | - Ninib Baryawno
- Center for Regenerative Medicine, Massachusetts General Hospital (MGH), Boston, Massachusetts, USA.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Naema Nayyar
- Center for Regenerative Medicine, Massachusetts General Hospital (MGH), Boston, Massachusetts, USA.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Yannis K Valtis
- Center for Regenerative Medicine, Massachusetts General Hospital (MGH), Boston, Massachusetts, USA.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Betty Yang
- Center for Regenerative Medicine, Massachusetts General Hospital (MGH), Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Ina Ly
- Department of Neurology and Division of Neuro-Oncology, MGH, and
| | - Antoine Besnard
- Center for Regenerative Medicine, Massachusetts General Hospital (MGH), Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Nicolas Severe
- Center for Regenerative Medicine, Massachusetts General Hospital (MGH), Boston, Massachusetts, USA.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Karin U Gustafsson
- Center for Regenerative Medicine, Massachusetts General Hospital (MGH), Boston, Massachusetts, USA.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Ovidiu C Andronesi
- Department of Radiology, Athinoula A. Martinos Biomedical Imaging Center, MGH, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Amar Sahay
- Center for Regenerative Medicine, Massachusetts General Hospital (MGH), Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - David T Scadden
- Center for Regenerative Medicine, Massachusetts General Hospital (MGH), Boston, Massachusetts, USA.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
126
|
Allogeneic bone marrow transplant in the absence of cytoreductive conditioning rescues mice with β-thalassemia major. Blood Adv 2017; 1:2421-2432. [PMID: 29296892 DOI: 10.1182/bloodadvances.2017009449] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 10/24/2017] [Indexed: 02/08/2023] Open
Abstract
β-thalassemia is a group of inherited blood disorders that result in defects in β-globin chain production. Cooley anemia (CA), or β-thalassemia major, is the most severe form of the disease and occurs when an individual has mutations in both copies of the adult β-globin gene. Patients with CA fail to make adult hemoglobin, exhibit ineffective erythropoiesis, experience severe anemia, and are transfusion dependent for life. Currently, allogeneic bone marrow transplantation (BMT) is the only cure; however, few patients have suitable donors for this procedure, which has significant morbidity and mortality. In this study, a novel humanized murine model of CA is rescued from lethal anemia by allogeneic BMT in the absence of cytoreductive conditioning. A single intravenous postnatal injection of allogeneic bone marrow results in stable, mixed hematopoietic chimerism. Five months after transplantation, donor cells accounted for approximately 90% of circulating erythrocytes and up to 15% of hematopoietic stem and progenitor cells. Transplanted mice are transfusion independent, have marked improvement of hematological indices, exhibit no growth retardation or signs of graft-versus-host disease, and are fertile. This study describes a method for the consistent engraftment of allogeneic donor hematopoietic cells that rescues a humanized mouse model of CA from lethal anemia, all in the absence of toxic cytoreductive conditioning.
Collapse
|
127
|
Nunes-Hasler P, Maschalidi S, Lippens C, Castelbou C, Bouvet S, Guido D, Bermont F, Bassoy EY, Page N, Merkler D, Hugues S, Martinvalet D, Manoury B, Demaurex N. STIM1 promotes migration, phagosomal maturation and antigen cross-presentation in dendritic cells. Nat Commun 2017; 8:1852. [PMID: 29176619 PMCID: PMC5701258 DOI: 10.1038/s41467-017-01600-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 10/02/2017] [Indexed: 12/31/2022] Open
Abstract
Antigen cross-presentation by dendritic cells (DC) stimulates cytotoxic T cell activation to promote immunity to intracellular pathogens, viruses and cancer. Phagocytosed antigens generate potent T cell responses, but the signalling and trafficking pathways regulating their cross-presentation are unclear. Here, we show that ablation of the store-operated-Ca2+-entry regulator STIM1 in mouse myeloid cells impairs cross-presentation and DC migration in vivo and in vitro. Stim1 ablation reduces Ca2+ signals, cross-presentation, and chemotaxis in mouse bone-marrow-derived DCs without altering cell differentiation, maturation or phagocytic capacity. Phagosomal pH homoeostasis and ROS production are unaffected by STIM1 deficiency, but phagosomal proteolysis and leucyl aminopeptidase activity, IRAP recruitment, as well as fusion of phagosomes with endosomes and lysosomes are all impaired. These data suggest that STIM1-dependent Ca2+ signalling promotes the delivery of endolysosomal enzymes to phagosomes to enable efficient cross-presentation. STIM proteins sense Ca2+ depletion in the ER and activate store-operated Ca2+-entry (SOCE) in response, a process associated with dendritic cell functions. Here the authors show STIM1 is the major isoform controlling SOCE in mouse dendritic cells and provide a mechanism for its requirement in antigen cross-presentation.
Collapse
Affiliation(s)
- Paula Nunes-Hasler
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, 1211, Switzerland.
| | - Sophia Maschalidi
- Laboratory of Normal and Pathological Homeostasis of the Immune System, INSERM UMR1163, Paris, 75015, France.,Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine Paris Descartes, Paris, 75015, France
| | - Carla Lippens
- Department of Pathology and Immunology, University of Geneva, Geneva, 1211, Switzerland
| | - Cyril Castelbou
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, 1211, Switzerland
| | - Samuel Bouvet
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, 1211, Switzerland
| | - Daniele Guido
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, 1211, Switzerland
| | - Flavien Bermont
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, 1211, Switzerland
| | - Esen Y Bassoy
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, 1211, Switzerland
| | - Nicolas Page
- Department of Pathology and Immunology, University of Geneva, Geneva, 1211, Switzerland
| | - Doron Merkler
- Department of Pathology and Immunology, University of Geneva, Geneva, 1211, Switzerland.,Division of Clinical Pathology, Geneva University Hospital, Geneva, 1211, Switzerland
| | - Stéphanie Hugues
- Department of Pathology and Immunology, University of Geneva, Geneva, 1211, Switzerland
| | - Denis Martinvalet
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, 1211, Switzerland
| | - Bénédicte Manoury
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de médecine Paris Descartes, Paris, 75015, France.,Institut National de la Santé et de la Recherche Médicale, Unité 1151, Paris, 75014, France.,Centre National de la Recherche Scientifique, Unité 8253, Paris, 75014, France
| | - Nicolas Demaurex
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, 1211, Switzerland
| |
Collapse
|
128
|
Gundry MC, Brunetti L, Lin A, Mayle AE, Kitano A, Wagner D, Hsu JI, Hoegenauer KA, Rooney CM, Goodell MA, Nakada D. Highly Efficient Genome Editing of Murine and Human Hematopoietic Progenitor Cells by CRISPR/Cas9. Cell Rep 2017; 17:1453-1461. [PMID: 27783956 DOI: 10.1016/j.celrep.2016.09.092] [Citation(s) in RCA: 196] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 07/01/2016] [Accepted: 09/28/2016] [Indexed: 12/17/2022] Open
Abstract
Our understanding of the mechanisms that regulate hematopoietic stem/progenitor cells (HSPCs) has been advanced by the ability to genetically manipulate mice; however, germline modification is time consuming and expensive. Here, we describe fast, efficient, and cost-effective methods to directly modify the genomes of mouse and human HSPCs using the CRISPR/Cas9 system. Using plasmid and virus-free delivery of guide RNAs alone into Cas9-expressing HSPCs or Cas9-guide RNA ribonucleoprotein (RNP) complexes into wild-type cells, we have achieved extremely efficient gene disruption in primary HSPCs from mouse (>60%) and human (∼75%). These techniques enabled rapid evaluation of the functional effects of gene loss of Eed, Suz12, and DNMT3A. We also achieved homology-directed repair in primary human HSPCs (>20%). These methods will significantly expand applications for CRISPR/Cas9 technologies for studying normal and malignant hematopoiesis.
Collapse
Affiliation(s)
- Michael C Gundry
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lorenzo Brunetti
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, 06156 Perugia, Italy
| | - Angelique Lin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Integrative Molecular and Biomedical Sciences Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Allison E Mayle
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ayumi Kitano
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Dimitrios Wagner
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; Texas Children's Hospital and Houston Methodist Hospital, Houston, TX 77030, USA; Institute for Medical Immunology, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Joanne I Hsu
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX 77030, USA; Translational Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kevin A Hoegenauer
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cliona M Rooney
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Texas Children's Hospital and Houston Methodist Hospital, Houston, TX 77030, USA
| | - Margaret A Goodell
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Texas Children's Hospital and Houston Methodist Hospital, Houston, TX 77030, USA.
| | - Daisuke Nakada
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
129
|
Granulocyte-derived TNFα promotes vascular and hematopoietic regeneration in the bone marrow. Nat Med 2017; 24:95-102. [PMID: 29155425 PMCID: PMC5760474 DOI: 10.1038/nm.4448] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 10/23/2017] [Indexed: 02/07/2023]
|
130
|
Hudson KE, de Wolski K, Kapp LM, Richards AL, Schniederjan MJ, Zimring JC. Antibodies to Senescent Antigen and C3 Are Not Required for Normal Red Blood Cell Lifespan in a Murine Model. Front Immunol 2017; 8:1425. [PMID: 29163500 PMCID: PMC5670101 DOI: 10.3389/fimmu.2017.01425] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 10/13/2017] [Indexed: 12/24/2022] Open
Abstract
Red blood cells (RBCs) have a well-defined lifespan, indicating a mechanism by which senescent cells of a certain age are removed from circulation. However, the specifics by which senescent cells are recognized and removed are poorly understood. There are multiple competing hypotheses for this process, perhaps the most commonly cited is that senescent RBCs expose neoantigens [or senescent antigen(s)] that are then recognized by naturally occurring antibodies, which opsonize the senescent cells and result in clearance from circulation. While there are a large volume of published data to indicate that older RBCs accumulate increased levels of antibody on their surface, to the best of our knowledge, the causal role of such antibodies in clearance has not been rigorously assessed. In the current report, we demonstrate that RBC lifespan and clearance patterns are not altered in mice deficient in antibodies, in C3 protein, or missing both. These data demonstrate that neither antibody nor C3 is required for clearance of senescent RBCs, and questions if they are even involved, in a murine model of RBC lifespan.
Collapse
Affiliation(s)
| | - Karen de Wolski
- Bloodworks Northwest Research Institute, Seattle, WA, United States
| | - Linda M Kapp
- Bloodworks Northwest Research Institute, Seattle, WA, United States
| | | | - Matthew J Schniederjan
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, United States
| | - James C Zimring
- Bloodworks Northwest Research Institute, Seattle, WA, United States.,Department of Laboratory Medicine, Division of Hematology, University of Washington, Seattle, WA, United States.,Department of Internal Medicine, Division of Hematology, University of Washington, Seattle, WA, United States
| |
Collapse
|
131
|
Mrass P, Oruganti SR, Fricke GM, Tafoya J, Byrum JR, Yang L, Hamilton SL, Miller MJ, Moses ME, Cannon JL. ROCK regulates the intermittent mode of interstitial T cell migration in inflamed lungs. Nat Commun 2017; 8:1010. [PMID: 29044117 PMCID: PMC5647329 DOI: 10.1038/s41467-017-01032-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 08/14/2017] [Indexed: 12/27/2022] Open
Abstract
Effector T cell migration through tissues can enable control of infection or mediate inflammatory damage. Nevertheless, the molecular mechanisms that regulate migration of effector T cells within the interstitial space of inflamed lungs are incompletely understood. Here, we show T cell migration in a mouse model of acute lung injury with two-photon imaging of intact lung tissue. Computational analysis indicates that T cells migrate with an intermittent mode, switching between confined and almost straight migration, guided by lung-associated vasculature. Rho-associated protein kinase (ROCK) is required for both high-speed migration and straight motion. By contrast, inhibition of Gαi signaling with pertussis toxin affects speed but not the intermittent migration of lung-infiltrating T cells. Computational modeling shows that an intermittent migration pattern balances both search area and the duration of contacts between T cells and target cells. These data identify that ROCK-dependent intermittent T cell migration regulates tissue-sampling during acute lung injury. ROCK is associated with T cell movement in lymph nodes. Here the authors use an LPS lung damage model and two-photon imaging to show that CD8+ T cells in lung tissue engage in ROCK-dependent fast linear migration alternating with bursts of slower confined migration that together optimize contact with target cells.
Collapse
Affiliation(s)
- Paulus Mrass
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, MSC 08 4660, 1 University of New Mexico, Albuquerque, NM, 87131, USA
| | - Sreenivasa Rao Oruganti
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, MSC 08 4660, 1 University of New Mexico, Albuquerque, NM, 87131, USA
| | - G Matthew Fricke
- Department of Computer Science, University of New Mexico, 1 University of New Mexico, Albuquerque, NM, 87131, USA
| | - Justyna Tafoya
- Department of Computer Science, University of New Mexico, 1 University of New Mexico, Albuquerque, NM, 87131, USA.,Department of Mathematics, University of New Mexico, 1 University of New Mexico, Albuquerque, NM, 87131, USA
| | - Janie R Byrum
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, MSC 08 4660, 1 University of New Mexico, Albuquerque, NM, 87131, USA
| | - Lihua Yang
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Samantha L Hamilton
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Mark J Miller
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Melanie E Moses
- Department of Computer Science, University of New Mexico, 1 University of New Mexico, Albuquerque, NM, 87131, USA.,Department of Biology, University of New Mexico, 1 University of New Mexico, Albuquerque, NM, 87131, USA.,External Faculty, Santa Fe Institute, 1399 Hyde Park Road, Santa Fe, NM, 87501, USA
| | - Judy L Cannon
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, MSC 08 4660, 1 University of New Mexico, Albuquerque, NM, 87131, USA.
| |
Collapse
|
132
|
Mohning MP, Thomas SM, Barthel L, Mould KJ, McCubbrey AL, Frasch SC, Bratton DL, Henson PM, Janssen WJ. Phagocytosis of microparticles by alveolar macrophages during acute lung injury requires MerTK. Am J Physiol Lung Cell Mol Physiol 2017; 314:L69-L82. [PMID: 28935638 DOI: 10.1152/ajplung.00058.2017] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Microparticles are a newly recognized class of mediators in the pathophysiology of lung inflammation and injury, but little is known about the factors that regulate their accumulation and clearance. The primary objective of our study was to determine whether alveolar macrophages engulf microparticles and to elucidate the mechanisms by which this occurs. Alveolar microparticles were quantified in bronchoalveolar fluid of mice with lung injury induced by LPS and hydrochloric acid. Microparticle numbers were greatest at the peak of inflammation and declined as inflammation resolved. Isolated, fluorescently labeled particles were placed in culture with macrophages to evaluate ingestion in the presence of endocytosis inhibitors. Ingestion was blocked with cytochalasin D and wortmannin, consistent with a phagocytic process. In separate experiments, mice were treated intratracheally with labeled microparticles, and their uptake was assessed though microscopy and flow cytometry. Resident alveolar macrophages, not recruited macrophages, were the primary cell-ingesting microparticles in the alveolus during lung injury. In vitro, microparticles promoted inflammatory signaling in LPS primed epithelial cells, signifying the importance of microparticle clearance in resolving lung injury. Microparticles were found to have phosphatidylserine exposed on their surfaces. Accordingly, we measured expression of phosphatidylserine receptors on macrophages and found high expression of MerTK and Axl in the resident macrophage population. Endocytosis of microparticles was markedly reduced in MerTK-deficient macrophages in vitro and in vivo. In conclusion, microparticles are released during acute lung injury and peak in number at the height of inflammation. Resident alveolar macrophages efficiently clear these microparticles through MerTK-mediated phagocytosis.
Collapse
Affiliation(s)
- Michael P Mohning
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health , Denver, Colorado.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Colorado-Anschutz Medical Campus , Aurora, Colorado
| | - Stacey M Thomas
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health , Denver, Colorado
| | - Lea Barthel
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health , Denver, Colorado
| | - Kara J Mould
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Colorado-Anschutz Medical Campus , Aurora, Colorado
| | - Alexandria L McCubbrey
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health , Denver, Colorado.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Colorado-Anschutz Medical Campus , Aurora, Colorado
| | | | - Donna L Bratton
- Department of Pediatrics, National Jewish Health , Denver, Colorado
| | - Peter M Henson
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Colorado-Anschutz Medical Campus , Aurora, Colorado.,Department of Pediatrics, National Jewish Health , Denver, Colorado
| | - William J Janssen
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health , Denver, Colorado.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Colorado-Anschutz Medical Campus , Aurora, Colorado
| |
Collapse
|
133
|
Abstract
During antibody affinity maturation, germinal center (GC) B cells cycle between affinity-driven selection in the light zone (LZ) and proliferation and somatic hypermutation in the dark zone (DZ). Although selection of GC B cells is triggered by antigen-dependent signals delivered in the LZ, DZ proliferation occurs in the absence of such signals. We show that positive selection triggered by T cell help activates the mechanistic target of rapamycin complex 1 (mTORC1), which promotes the anabolic program that supports DZ proliferation. Blocking mTORC1 prior to growth prevented clonal expansion, whereas blockade after cells reached peak size had little to no effect. Conversely, constitutively active mTORC1 led to DZ enrichment but loss of competitiveness and impaired affinity maturation. Thus, mTORC1 activation is required for fueling B cells prior to DZ proliferation rather than for allowing cell-cycle progression itself and must be regulated dynamically during cyclic re-entry to ensure efficient affinity-based selection.
Collapse
|
134
|
Johnson V, Webb T, Norman A, Coy J, Kurihara J, Regan D, Dow S. Activated Mesenchymal Stem Cells Interact with Antibiotics and Host Innate Immune Responses to Control Chronic Bacterial Infections. Sci Rep 2017; 7:9575. [PMID: 28851894 PMCID: PMC5575141 DOI: 10.1038/s41598-017-08311-4] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 06/19/2017] [Indexed: 12/26/2022] Open
Abstract
Chronic bacterial infections associated with biofilm formation are often difficult to resolve without extended courses of antibiotic therapy. Mesenchymal stem cells (MSC) exert antibacterial activity in vitro and in acute bacterial infection models, but their activity in chronic infection with biofilm models has not been previously investigated. Therefore, we studied the effects of MSC administration in mouse and dog models of chronic infections associated with biofilms. Mice with chronic Staphylococcus aureus implant infections were treated by i.v. administration of activated or non-activated MSC, with or without antibiotic therapy. The most effective treatment protocol was identified as activated MSC co-administered with antibiotic therapy. Activated MSC were found to accumulate in the wound margins several days after i.v. administration. Macrophages in infected tissues assumed an M2 phenotype, compared to untreated infections which contained predominately M1 macrophages. Bacterial killing by MSC was found to be mediated in part by secretion of cathelicidin and was significantly increased by antibiotics. Studies in pet dogs with spontaneous chronic multi drug-resistant wound infections demonstrated clearance of bacteria and wound healing following repeated i.v. administration of activated allogeneic canine MSC. Thus, systemic therapy with activated MSC may be an effective new, non-antimicrobial approach to treatment of chronic, drug-resistant infections.
Collapse
Affiliation(s)
- Valerie Johnson
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Ft. Collins, CO, USA
| | - Tracy Webb
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Ft. Collins, CO, USA
| | - Annalis Norman
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Ft. Collins, CO, USA
| | - Jonathan Coy
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Ft. Collins, CO, USA
| | - Jade Kurihara
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Ft. Collins, CO, USA
| | - Daniel Regan
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Ft. Collins, CO, USA
| | - Steven Dow
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Ft. Collins, CO, USA.
| |
Collapse
|
135
|
Rizzi N, Rebecchi M, Levandis G, Ciana P, Maggi A. Identification of novel loci for the generation of reporter mice. Nucleic Acids Res 2017; 45:e37. [PMID: 27899606 PMCID: PMC5389565 DOI: 10.1093/nar/gkw1142] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 11/08/2016] [Indexed: 12/25/2022] Open
Abstract
Deciphering the etiology of complex pathologies at molecular level requires longitudinal studies encompassing multiple biochemical pathways (apoptosis, proliferation, inflammation, oxidative stress). In vivo imaging of current reporter animals enabled the spatio-temporal analysis of specific molecular events, however, the lack of a multiplicity of loci for the generalized and regulated expression of the integrated transgenes hampers the creation of systems for the simultaneous analysis of more than a biochemical pathways at the time. We here developed and tested an in vivo-based methodology for the identification of multiple insertional loci suitable for the generation of reliable reporter mice. The validity of the methodology was tested with the generation of novel mice useful to report on inflammation and oxidative stress.
Collapse
Affiliation(s)
- Nicoletta Rizzi
- Center of Excellence on Neurodegenerative Diseases and Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9 20133 Milan, Italy
| | - Monica Rebecchi
- Center of Excellence on Neurodegenerative Diseases and Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9 20133 Milan, Italy
| | - Giovanna Levandis
- Center of Excellence on Neurodegenerative Diseases and Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9 20133 Milan, Italy
| | - Paolo Ciana
- Center of Excellence on Neurodegenerative Diseases and Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Via Balzaretti 9 20133 Milan, Italy
| | - Adriana Maggi
- Center of Excellence on Neurodegenerative Diseases and Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9 20133 Milan, Italy
| |
Collapse
|
136
|
Berry MR, Mathews RJ, Ferdinand JR, Jing C, Loudon KW, Wlodek E, Dennison TW, Kuper C, Neuhofer W, Clatworthy MR. Renal Sodium Gradient Orchestrates a Dynamic Antibacterial Defense Zone. Cell 2017; 170:860-874.e19. [DOI: 10.1016/j.cell.2017.07.022] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 06/17/2017] [Accepted: 07/14/2017] [Indexed: 12/24/2022]
|
137
|
Local lung hypoxia determines epithelial fate decisions during alveolar regeneration. Nat Cell Biol 2017; 19:904-914. [PMID: 28737769 PMCID: PMC5600325 DOI: 10.1038/ncb3580] [Citation(s) in RCA: 200] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 06/21/2017] [Indexed: 12/12/2022]
Abstract
After influenza infection, lineage-negative epithelial progenitors (LNEPs) exhibit a binary response to reconstitute epithelial barriers: activating a Notch-dependent ΔNp63/cytokeratin 5 (Krt5) remodelling program or differentiating into alveolar type II cells (AEC2s). Here we show that local lung hypoxia, through hypoxia-inducible factor (HIF1α), drives Notch signalling and Krt5pos basal-like cell expansion. Single-cell transcriptional profiling of human AEC2s from fibrotic lungs revealed a hypoxic subpopulation with activated Notch, suppressed surfactant protein C (SPC), and transdifferentiation toward a Krt5pos basal-like state. Activated murine Krt5pos LNEPs and diseased human AEC2s upregulate strikingly similar core pathways underlying migration and squamous metaplasia. While robust, HIF1α-driven metaplasia is ultimately inferior to AEC2 reconstitution in restoring normal lung function. HIF1α deletion or enhanced Wnt/β-catenin activity in Sox2pos LNEPs blocks Notch and Krt5 activation, instead promoting rapid AEC2 differentiation and migration and improving the quality of alveolar repair.
Collapse
|
138
|
The BH3-only proteins BIM and PUMA are not critical for the reticulocyte apoptosis caused by loss of the pro-survival protein BCL-XL. Cell Death Dis 2017; 8:e2914. [PMID: 28682312 PMCID: PMC5550852 DOI: 10.1038/cddis.2017.304] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 05/02/2017] [Indexed: 01/06/2023]
Abstract
Anaemia is a major global health problem arising from diverse causes and for which improved therapeutic strategies are needed. Erythroid cells can undergo apoptotic cell death and loss of pro-survival BCL-XL is known to trigger apoptosis during late-stage erythroid development. However, the mechanism by which loss or pharmacological blockade of BCL-XL leads to erythroid cell apoptosis remains unclear. Here we sought to identify the precise stage of erythropoiesis that depends on BCL-XL. We also tested whether deficiency of BIM or PUMA, the two main pro-apoptotic antagonists of BCL-XL, could prevent reticulocyte death and anaemia caused by BCL-XL loss. Using an in vivo mouse model of tamoxifen-inducible Bclx gene deletion and in vitro assays with a BCL-XL-selective inhibitor, we interrogated each stage of erythrocyte differentiation for BCL-XL dependency. This revealed that reticulocytes, but not orthochromatic erythroblasts, require BCL-XL for their survival. Surprisingly, concurrent loss of BIM or PUMA had no significant impact on the development of anemia following acute BCL-XL deletion in vivo. However, analysis of mixed bone marrow chimaeric mice revealed that loss of PUMA, but not loss of BIM, partially alleviated impaired erythropoiesis caused by BCL-XL deficiency. Insight into how the network of pro-survival and pro-apoptotic proteins works will assist the development of strategies to mitigate the effects of abnormal cell death during erythropoiesis and prevent anaemia in patients treated with BCL-XL-specific BH3-mimetic drugs.
Collapse
|
139
|
Lymphatic endothelial S1P promotes mitochondrial function and survival in naive T cells. Nature 2017; 546:158-161. [PMID: 28538737 PMCID: PMC5683179 DOI: 10.1038/nature22352] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 04/07/2017] [Indexed: 02/07/2023]
|
140
|
Carneiro MB, Hohman LS, Egen JG, Peters NC. Use of two-photon microscopy to study Leishmania major infection of the skin. Methods 2017; 127:45-52. [PMID: 28434998 DOI: 10.1016/j.ymeth.2017.04.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/21/2017] [Accepted: 04/12/2017] [Indexed: 10/19/2022] Open
Abstract
Intra-vital two-photon microscopy (2P-IVM) allows for in-situ investigation of tissue organization, cell behavior and the dynamic interactions between different cell types in their natural environment. This methodology has also expanded our understanding of the immune response against pathogens. Leishmania are protozoan intracellular parasites that have adapted to successfully establish infection within the context of an inflammatory response in the skin following transmission by the bite of an infected sand fly. The generation of fluorescent transgenic parasites coupled with the increased availability of different types of fluorescent transgenic reporter mice has facilitated the study of the host-parasite interaction in the skin, significantly impacting our understanding of cutaneous leishmaniasis. In this review we will discuss 2P-IVM in the context of Leishmania infection of the mouse ear skin and describe a simple and minimally invasive procedure that allows long-term imaging of this host-pathogen interaction.
Collapse
Affiliation(s)
- Matheus Batista Carneiro
- Snyder Institute for Chronic Diseases, Departments of Microbiology, Immunology and Infectious Disease, Cumming School of Medicine and Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, AB T2N 4Z6, Canada.
| | - Leah Shan Hohman
- Snyder Institute for Chronic Diseases, Departments of Microbiology, Immunology and Infectious Disease, Cumming School of Medicine and Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, AB T2N 4Z6, Canada
| | - Jackson G Egen
- Department of Oncology Research, Amgen Inc, South San Francisco, CA 94080, USA
| | - Nathan C Peters
- Snyder Institute for Chronic Diseases, Departments of Microbiology, Immunology and Infectious Disease, Cumming School of Medicine and Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, AB T2N 4Z6, Canada.
| |
Collapse
|
141
|
Haghayegh Jahromi N, Tardent H, Enzmann G, Deutsch U, Kawakami N, Bittner S, Vestweber D, Zipp F, Stein JV, Engelhardt B. A Novel Cervical Spinal Cord Window Preparation Allows for Two-Photon Imaging of T-Cell Interactions with the Cervical Spinal Cord Microvasculature during Experimental Autoimmune Encephalomyelitis. Front Immunol 2017; 8:406. [PMID: 28443093 PMCID: PMC5387098 DOI: 10.3389/fimmu.2017.00406] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 03/22/2017] [Indexed: 11/13/2022] Open
Abstract
T-cell migration across the blood-brain barrier (BBB) is a crucial step in the pathogenesis of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). Two-photon intravital microscopy (2P-IVM) has been established as a powerful tool to study cell-cell interactions in inflammatory EAE lesions in living animals. In EAE, central nervous system inflammation is strongly pronounced in the spinal cord, an organ in which 2P-IVM imaging is technically very challenging and has been limited to the lumbar spinal cord. Here, we describe a novel spinal cord window preparation allowing to use 2P-IVM to image immune cell interactions with the cervical spinal cord microvascular endothelium during EAE. We describe differences in the angioarchitecture of the cervical spinal cord versus the lumbar spinal cord, which will entail different hemodynamic parameters in these different vascular beds. Using T cells as an example, we demonstrate the suitability of this novel methodology in imaging the post-arrest multistep T-cell extravasation across the cervical spinal cord microvessels. The novel methodology includes an outlook to the analysis of the cellular pathway of T-cell diapedesis across the BBB by establishing visualization of endothelial junctions in this vascular bed.
Collapse
Affiliation(s)
| | - Heidi Tardent
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Gaby Enzmann
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Urban Deutsch
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Naoto Kawakami
- Max Planck Institute of Neurobiology, Martinsried, Germany.,Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians University of Munich, Martinsried, Germany
| | - Stefan Bittner
- Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | | | - Frauke Zipp
- Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Jens V Stein
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | | |
Collapse
|
142
|
McNamara HA, Cai Y, Wagle MV, Sontani Y, Roots CM, Miosge LA, O'Connor JH, Sutton HJ, Ganusov VV, Heath WR, Bertolino P, Goodnow CG, Parish IA, Enders A, Cockburn IA. Up-regulation of LFA-1 allows liver-resident memory T cells to patrol and remain in the hepatic sinusoids. Sci Immunol 2017; 2. [PMID: 28707003 DOI: 10.1126/sciimmunol.aaj1996] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Liver-resident CD8+ T cells are highly motile cells that patrol the vasculature and provide protection against liver pathogens. A key question is: how can these liver CD8+ T cells be simultaneously present in the circulation and tissue-resident? Because liver-resident T cells do not express CD103 - a key integrin for T cell residence in epithelial tissues - we investigated other candidate adhesion molecules. Using intra-vital imaging we found that CD8+ T cell patrolling in the hepatic sinusoids is dependent upon LFA-1-ICAM-1 interactions. Interestingly, liver-resident CD8+ T cells up-regulate LFA-1 compared to effector-memory cells, presumably to facilitate this behavior. Finally, we found that LFA-1 deficient CD8+ T cells failed to form substantial liver-resident memory populations following Plasmodium or LCMV immunization. Collectively, our results demonstrate that it is adhesion through LFA-1 that allows liver-resident memory CD8+ T cells to patrol and remain in the hepatic sinusoids.
Collapse
Affiliation(s)
- H A McNamara
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2602, Australia
| | - Y Cai
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2602, Australia
| | - M V Wagle
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2602, Australia
| | - Y Sontani
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2602, Australia
| | - C M Roots
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2602, Australia
| | - L A Miosge
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2602, Australia
| | - J H O'Connor
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2602, Australia
| | - H J Sutton
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2602, Australia
| | - V V Ganusov
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, United States of America
| | - W R Heath
- Department of Microbiology and Immunology, The Peter Doherty Institute, University of Melbourne, Parkville, VIC 3010, Australia
| | - P Bertolino
- Liver Immunology Program, Centenary Institute and AW Morrow Gastroenterology and Liver Centre, University of Sydney and Royal Prince Alfred Hospital, Locked Bag No. 6, Sydney, NSW 2042, Australia
| | - C G Goodnow
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2602, Australia.,Immunogenomics Laboratory, Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW 2010, Australia
| | - I A Parish
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2602, Australia
| | - A Enders
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2602, Australia
| | - I A Cockburn
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2602, Australia
| |
Collapse
|
143
|
Lippens C, Duraes FV, Dubrot J, Brighouse D, Lacroix M, Irla M, Aubry-Lachainaye JP, Reith W, Mandl JN, Hugues S. IDO-orchestrated crosstalk between pDCs and Tregs inhibits autoimmunity. J Autoimmun 2016; 75:39-49. [PMID: 27470005 PMCID: PMC5127883 DOI: 10.1016/j.jaut.2016.07.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 07/07/2016] [Accepted: 07/10/2016] [Indexed: 01/21/2023]
Abstract
Plasmacytoid dendritic cells (pDCs) have been shown to both mediate and prevent autoimmunity, and the regulation of their immunogenic versus tolerogenic functions remains incompletely understood. Here we demonstrate that, compared to other cells, pDCs are the major expressors of Indoleamine-2,3-dioxygenase (IDO) in steady-state lymph nodes (LNs). IDO expression by LN pDCs was closely dependent on MHCII-mediated, antigen-dependent, interactions with Treg. We further established that IDO production by pDCs was necessary to confer suppressive function to Tregs. During EAE development, IDO expression by pDCs was required for the generation of Tregs capable of dampening the priming of encephalitogenic T cell and disease severity. Thus, we describe a novel crosstalk between pDCs and Tregs: Tregs shape tolerogenic functions of pDCs prior to inflammation, such that pDCs in turn, promote Treg suppressive functions during autoimmunity.
Collapse
MESH Headings
- Animals
- Autoimmunity/genetics
- Autoimmunity/immunology
- Cells, Cultured
- Coculture Techniques
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Encephalomyelitis, Autoimmune, Experimental/enzymology
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Flow Cytometry
- Gene Expression Regulation, Enzymologic
- Histocompatibility Antigens Class II/immunology
- Histocompatibility Antigens Class II/metabolism
- Humans
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Lymph Nodes/enzymology
- Lymph Nodes/immunology
- Mice, Inbred C57BL
- Mice, Transgenic
- Reverse Transcriptase Polymerase Chain Reaction
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
Collapse
Affiliation(s)
- Carla Lippens
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Fernanda V Duraes
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Juan Dubrot
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Dale Brighouse
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Mathilde Lacroix
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Magali Irla
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | | | - Walter Reith
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Judith N Mandl
- Lymphocyte Biology Section, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Stéphanie Hugues
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland.
| |
Collapse
|
144
|
Gradients of the signaling lipid S1P in lymph nodes position natural killer cells and regulate their interferon-γ response. Nat Immunol 2016; 18:15-25. [PMID: 27841869 DOI: 10.1038/ni.3619] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 10/20/2016] [Indexed: 12/14/2022]
Abstract
The lymph node periphery is an important site for many immunological functions, from pathogen containment to the differentiation of helper T cells, yet the cues that position cells in this region are largely undefined. Here, through the use of a reporter for the signaling lipid S1P (sphingosine 1-phosphate), we found that cells sensed higher concentrations of S1P in the medullary cords than in the T cell zone and that the S1P transporter SPNS2 on lymphatic endothelial cells generated this gradient. Natural killer (NK) cells are located at the periphery of the lymph node, predominantly in the medulla, and we found that expression of SPNS2, expression of the S1P receptor S1PR5 on NK cells, and expression of the chemokine receptor CXCR4 were all required for NK cell localization during homeostasis and rapid production of interferon-γ by NK cells after challenge. Our findings elucidate the spatial cues for NK cell organization and reveal a previously unknown role for S1P in positioning cells within the medulla.
Collapse
|
145
|
Fallet B, Narr K, Ertuna YI, Remy M, Sommerstein R, Cornille K, Kreutzfeldt M, Page N, Zimmer G, Geier F, Straub T, Pircher H, Larimore K, Greenberg PD, Merkler D, Pinschewer DD. Interferon-driven deletion of antiviral B cells at the onset of chronic infection. Sci Immunol 2016; 1:eaah6817. [PMID: 27872905 PMCID: PMC5115616 DOI: 10.1126/sciimmunol.aah6817] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Inadequate antibody responses and perturbed B cell compartments represent hallmarks of persistent microbial infections, but the mechanisms whereby persisting pathogens suppress humoral immunity remain poorly defined. Using adoptive transfer experiments in the context of a chronic lymphocytic choriomeningitis virus (LCMV) infection of mice, we have documented rapid depletion of virus-specific B cells that coincided with the early type I interferon response to infection. We found that the loss of activated B cells was driven by type I interferon (IFN-I) signaling to several cell types including dendritic cells, T cells and myeloid cells. Intriguingly, this process was independent of B cell-intrinsic IFN-I sensing and resulted from biased differentiation of naïve B cells into short-lived antibody-secreting cells. The ability to generate robust B cell responses was restored upon IFN-I receptor blockade or, partially, when experimentally depleting myeloid cells or the IFN-I-induced cytokines interleukin 10 and tumor necrosis factor alpha. We have termed this IFN-I-driven depletion of B cells "B cell decimation". Strategies to counter "B cell decimation" should thus help us better leverage humoral immunity in the combat against persistent microbial diseases.
Collapse
Affiliation(s)
- Benedict Fallet
- Department of Biomedicine, Division of Experimental Virology, University of Basel, 4003 Basel, Switzerland
| | - Kerstin Narr
- Department of Biomedicine, Division of Experimental Virology, University of Basel, 4003 Basel, Switzerland
| | - Yusuf I. Ertuna
- Department of Biomedicine, Division of Experimental Virology, University of Basel, 4003 Basel, Switzerland
| | - Melissa Remy
- Department of Biomedicine, Division of Experimental Virology, University of Basel, 4003 Basel, Switzerland
| | - Rami Sommerstein
- Department of Pathology and Immunology, Geneva Faculty of Medicine, 1211 Geneva 4, Switzerland
| | - Karen Cornille
- Department of Biomedicine, Division of Experimental Virology, University of Basel, 4003 Basel, Switzerland
| | - Mario Kreutzfeldt
- Department of Pathology and Immunology, Geneva Faculty of Medicine, 1211 Geneva 4, Switzerland
- Division of Clinical Pathology, University Hospital Geneva, 1 rue Michel Servet, 1211 Geneva 4, Switzerland
| | - Nicolas Page
- Department of Pathology and Immunology, Geneva Faculty of Medicine, 1211 Geneva 4, Switzerland
| | - Gert Zimmer
- Institute of Virology and Immunology IVI, 3147 Mittelhäusern, Switzerland
| | - Florian Geier
- Department of Biomedicine, Bioinformatics Core Facility, University Hospital Basel, 4031 Basel, Switzerland
| | - Tobias Straub
- Institute for Immunology, Department for Medical Microbiology and Hygiene, University Medical Center Freiburg, 79104 Freiburg, Germany
| | - Hanspeter Pircher
- Institute for Immunology, Department for Medical Microbiology and Hygiene, University Medical Center Freiburg, 79104 Freiburg, Germany
| | - Kevin Larimore
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA 98109, USA
- Department of Immunology, University of Washington, Seattle, Washington, WA 98109, USA
| | - Philip D. Greenberg
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA 98109, USA
- Department of Immunology, University of Washington, Seattle, Washington, WA 98109, USA
| | - Doron Merkler
- Department of Pathology and Immunology, Geneva Faculty of Medicine, 1211 Geneva 4, Switzerland
- Division of Clinical Pathology, University Hospital Geneva, 1 rue Michel Servet, 1211 Geneva 4, Switzerland
| | - Daniel D. Pinschewer
- Department of Biomedicine, Division of Experimental Virology, University of Basel, 4003 Basel, Switzerland
| |
Collapse
|
146
|
Anti-MMP-9 Antibody: A Promising Therapeutic Strategy for Treatment of Inflammatory Bowel Disease Complications with Fibrosis. Inflamm Bowel Dis 2016; 22:2041-57. [PMID: 27542125 DOI: 10.1097/mib.0000000000000863] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND Despite medical treatments or surgical options, more than one-third of patients with Crohn's disease suffer from recurring fistulae. Matrix metalloprotease 9 (MMP-9), a type IV collagenase that cleaves components of the extracellular matrix leading to tissue remodeling, is upregulated in crypt abscesses and around fistulae suggesting an important role for this enzyme in fistula formation. Our aims were (1) to correlate serum levels of MMP-9 degradation products in patients with CD with the presence of fistulae and (2) to investigate the impact of selective MMP-9 inhibition in a mouse model of intestinal fibrosis. METHODS Serum MMP-9 degradation products were quantified in subjects affected with nonstricturing and nonpenetrating CD (n = 50), stricturing CD (n = 41), penetrating CD (n = 22), CD with perianal fistula (n = 29), and healthy controls (n = 10). Therapeutic efficacy of anti-MMP-9 monoclonal antibodies was assessed in a heterotopic xenograft model of intestinal fibrosis. RESULTS C3M, an MMP-9 degradation product of collagen III, demonstrated the highest serum levels in patients with penetrating CD and differentiated penetrating CD from other CD subgroups and healthy controls, P = 0.0005. Anti-MMP-9 treatments reduced collagen deposition and hydroxyproline content in day-14 intestinal grafts indicating reduced fibrosis. CONCLUSIONS The serologic biomarker C3M can discriminate penetrating CD from other CD subgroups and could serve as marker for the development of penetrating CD. Anti-MMP-9 antibody has therapeutic potential to prevent intestinal fibrosis in CD complications.
Collapse
|
147
|
Prather GR, MacLean JA, Shi M, Boadu DK, Paquet M, Hayashi K. Niclosamide As a Potential Nonsteroidal Therapy for Endometriosis That Preserves Reproductive Function in an Experimental Mouse Model. Biol Reprod 2016; 95:76. [PMID: 27535961 PMCID: PMC5333938 DOI: 10.1095/biolreprod.116.140236] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 08/16/2016] [Indexed: 01/05/2023] Open
Abstract
Endometriosis causes severe chronic pelvic pain and infertility. Because the standard medication and surgical treatments of endometriosis show high recurrence of symptoms, it is necessary to improve current treatment options. In the initial study, we examined whether niclosamide can be a useful drug for endometriosis in a preclinical setting. Endometriotic implants were induced using an established mouse model involving transimplantation of mouse endometrial fragments to the peritoneal wall of recipient mice. When the recipient mice were treated with niclosamide for 3 weeks, niclosamide reduced the size of endometriotic implants with inhibition of cell proliferation, and inflammatory signaling including RELA (NFKB) and STAT3 activation, but did not alter expression of steroid hormone receptors. To identify genes whose expression is regulated by niclosamide in endometriotic implants, RNA-sequencing was performed, and several genes downregulated by niclosamide were related to inflammatory responses, WNT and MAPK signaling. In a second study designed to assess whether niclosamide affects reproductive function, the recipient mice started receiving niclosamide after the induction of endometriosis. Then, the recipient mice were mated with wild type males, and treatments continued until the pups were born. Niclosamide treated recipient mice became pregnant and produced normal size and number of pups. These results suggest that niclosamide could be an effective therapeutic drug, and acts as an inhibitor of inflammatory signaling without disrupting normal reproductive function.
Collapse
Affiliation(s)
- Genna R. Prather
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, Illinois
| | - James A. MacLean
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, Illinois
| | - Mingxin Shi
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, Illinois
| | - Daniel K. Boadu
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, Illinois
| | - Marilène Paquet
- Departement de Pathologie et de Microbiologie, Université de Montreal, St-Hyacinthe, Quebec, Canada
| | - Kanako Hayashi
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, Illinois
- Correspondence: Kanako Hayashi, Department of Physiology, Southern Illinois University School of Medicine, 1135 Lincoln Dr., Carbondale, IL 62901. E-mail:
| |
Collapse
|
148
|
Sood A, Dong M, Melichar HJ. Preparation and Applications of Organotypic Thymic Slice Cultures. J Vis Exp 2016. [PMID: 27585240 DOI: 10.3791/54355] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Thymic selection proceeds in a unique and highly organized thymic microenvironment resulting in the generation of a functional, self-tolerant T cell repertoire. In vitro models to study T lineage commitment and development have provided valuable insights into this process. However, these systems lack the complete three-dimensional thymic milieu necessary for T cell development and, therefore, are incomplete approximations of in vivo thymic selection. Some of the challenges related to modeling T cell development can be overcome by using in situ models that provide an intact thymic microenvironment that fully supports thymic selection of developing T cells. Thymic slice organotypic cultures complement existing in situ techniques. Thymic slices preserve the integrity of the thymic cortical and medullary regions and provide a platform to study development of overlaid thymocytes of a defined developmental stage or of endogenous T cells within a mature thymic microenvironment. Given the ability to generate ~20 slices per mouse, thymic slices present a unique advantage in terms of scalability for high throughput experiments. Further, the relative ease in generating thymic slices and potential to overlay different thymic subsets or other cell populations from diverse genetic backgrounds enhances the versatility of this method. Here we describe a protocol for the preparation of thymic slices, isolation and overlay of thymocytes, and dissociation of thymic slices for flow cytometric analysis. This system can also be adapted to study non-conventional T cell development as well as visualize thymocyte migration, thymocyte-stromal cell interactions, and TCR signals associated with thymic selection by two-photon microscopy.
Collapse
Affiliation(s)
- Aditi Sood
- Centre de Recherche, Hôpital Maisonneuve-Rosemont; Department of Microbiology, Infectiology and Immunology, Université de Montréal
| | - Mengqi Dong
- Centre de Recherche, Hôpital Maisonneuve-Rosemont; Department of Microbiology, Infectiology and Immunology, Université de Montréal
| | - Heather J Melichar
- Centre de Recherche, Hôpital Maisonneuve-Rosemont; Department of Medicine, Université de Montréal;
| |
Collapse
|
149
|
Shimizu T, Kubovcakova L, Nienhold R, Zmajkovic J, Meyer SC, Hao-Shen H, Geier F, Dirnhofer S, Guglielmelli P, Vannucchi AM, Feenstra JDM, Kralovics R, Orkin SH, Skoda RC. Loss of Ezh2 synergizes with JAK2-V617F in initiating myeloproliferative neoplasms and promoting myelofibrosis. J Exp Med 2016; 213:1479-96. [PMID: 27401344 PMCID: PMC4986524 DOI: 10.1084/jem.20151136] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 06/16/2016] [Indexed: 12/29/2022] Open
Abstract
Skoda et al. provide new insights into the collaboration between epigenetic regulator Ezh2 and a key hematopoietic tyrosine kinase in disease initiation and progression. Myeloproliferative neoplasm (MPN) patients frequently show co-occurrence of JAK2-V617F and mutations in epigenetic regulator genes, including EZH2. In this study, we show that JAK2-V617F and loss of Ezh2 in hematopoietic cells contribute synergistically to the development of MPN. The MPN phenotype induced by JAK2-V617F was accentuated in JAK2-V617F;Ezh2−/− mice, resulting in very high platelet and neutrophil counts, more advanced myelofibrosis, and reduced survival. These mice also displayed expansion of the stem cell and progenitor cell compartments and a shift of differentiation toward megakaryopoiesis at the expense of erythropoiesis. Single cell limiting dilution transplantation with bone marrow from JAK2-V617F;Ezh2+/− mice showed increased reconstitution and MPN disease initiation potential compared with JAK2-V617F alone. RNA sequencing in Ezh2-deficient hematopoietic stem cells (HSCs) and megakaryocytic erythroid progenitors identified highly up-regulated genes, including Lin28b and Hmga2, and chromatin immunoprecipitation (ChIP)–quantitative PCR (qPCR) analysis of their promoters revealed decreased H3K27me3 deposition. Forced expression of Hmga2 resulted in increased chimerism and platelet counts in recipients of retrovirally transduced HSCs. JAK2-V617F–expressing mice treated with an Ezh2 inhibitor showed higher platelet counts than vehicle controls. Our data support the proposed tumor suppressor function of EZH2 in patients with MPN and call for caution when considering using Ezh2 inhibitors in MPN.
Collapse
Affiliation(s)
- Takafumi Shimizu
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| | - Lucia Kubovcakova
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| | - Ronny Nienhold
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| | - Jakub Zmajkovic
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| | - Sara C Meyer
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| | - Hui Hao-Shen
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| | - Florian Geier
- Bioinformatics Core Facility, Department of Biomedicine, University Hospital Basel, 4031 Basel, Switzerland
| | - Stephan Dirnhofer
- Institute of Pathology, University Hospital Basel, 4031 Basel, Switzerland
| | - Paola Guglielmelli
- Department of Clinical and Experimental Medicine, University of Florence, 50134 Florence, Italy
| | - Alessandro M Vannucchi
- Department of Clinical and Experimental Medicine, University of Florence, 50134 Florence, Italy
| | | | - Robert Kralovics
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Stuart H Orkin
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115 Department of Pediatric Oncology, Dana Farber Cancer Institute, Boston, MA 02215 Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02215
| | - Radek C Skoda
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| |
Collapse
|
150
|
Cabinian A, Sinsimer D, Tang M, Zumba O, Mehta H, Toma A, Sant’Angelo D, Laouar Y, Laouar A. Transfer of Maternal Immune Cells by Breastfeeding: Maternal Cytotoxic T Lymphocytes Present in Breast Milk Localize in the Peyer's Patches of the Nursed Infant. PLoS One 2016; 11:e0156762. [PMID: 27285085 PMCID: PMC4902239 DOI: 10.1371/journal.pone.0156762] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 05/19/2016] [Indexed: 12/22/2022] Open
Abstract
Despite our knowledge of the protective role of antibodies passed to infants through breast milk, our understanding of immunity transfer via maternal leukocytes is still limited. To emulate the immunological interface between the mother and her infant while breast-feeding, we used murine pups fostered after birth onto MHC-matched and MHC-mismatched dams. Overall, data revealed that: 1) Survival of breast milk leukocytes in suckling infants is possible, but not significant after the foster-nursing ceases; 2) Most breast milk lymphocytes establish themselves in specific areas of the intestine termed Peyer’s patches (PPs); 3) While most leukocytes in the milk bolus were myeloid cells, the majority of breast milk leukocytes localized to PPs were T lymphocytes, and cytotoxic T cells (CTLs) in particular; 4) These CTLs exhibit high levels of the gut-homing molecules α4β7 and CCR9, but a reduced expression of the systemic homing marker CD62L; 5) Under the same activation conditions, transferred CD8 T cells through breast milk have a superior capacity to produce potent cytolytic and inflammatory mediators when compared to those generated by the breastfed infant. It is therefore possible that maternal CTLs found in breast milk are directed to the PPs to compensate for the immature adaptive immune system of the infant in order to protect it against constant oral infectious risks during the postnatal phase.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Animals, Suckling
- Cells, Cultured
- Chemotaxis, Leukocyte/physiology
- Female
- Immunity, Maternally-Acquired/immunology
- Immunization, Passive/methods
- Lactation/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Transgenic
- Milk/cytology
- Milk/immunology
- Mothers
- Peyer's Patches/cytology
- Peyer's Patches/immunology
- T-Lymphocytes, Cytotoxic/cytology
- T-Lymphocytes, Cytotoxic/physiology
Collapse
Affiliation(s)
- Allison Cabinian
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, United States of America
| | - Daniel Sinsimer
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, United States of America
| | - May Tang
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, United States of America
| | - Osvaldo Zumba
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, United States of America
| | - Hetali Mehta
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, United States of America
| | - Annmarie Toma
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, United States of America
| | - Derek Sant’Angelo
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, United States of America
| | - Yasmina Laouar
- Department of Microbiology and Immunology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
- * E-mail: (AL); (YL)
| | - Amale Laouar
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, United States of America
- * E-mail: (AL); (YL)
| |
Collapse
|