Your browser doesn't support javascript.
loading
CSF1R-dependent macrophages control postnatal somatic growth and organ maturation.
Keshvari, Sahar; Caruso, Melanie; Teakle, Ngari; Batoon, Lena; Sehgal, Anuj; Patkar, Omkar L; Ferrari-Cestari, Michelle; Snell, Cameron E; Chen, Chen; Stevenson, Alex; Davis, Felicity M; Bush, Stephen J; Pridans, Clare; Summers, Kim M; Pettit, Allison R; Irvine, Katharine M; Hume, David A.
Afiliação
  • Keshvari S; Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Qld, Australia.
  • Caruso M; Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Qld, Australia.
  • Teakle N; Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Qld, Australia.
  • Batoon L; Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Qld, Australia.
  • Sehgal A; Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Qld, Australia.
  • Patkar OL; Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Qld, Australia.
  • Ferrari-Cestari M; Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Qld, Australia.
  • Snell CE; Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Qld, Australia.
  • Chen C; School of Biomedical Sciences, University of Queensland, St Lucia, Qld, Australia.
  • Stevenson A; Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Qld, Australia.
  • Davis FM; Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Qld, Australia.
  • Bush SJ; Nuffield Department of Clinical Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom.
  • Pridans C; Centre for Inflammation Research and Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom.
  • Summers KM; Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Qld, Australia.
  • Pettit AR; Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Qld, Australia.
  • Irvine KM; Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Qld, Australia.
  • Hume DA; Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Qld, Australia.
PLoS Genet ; 17(6): e1009605, 2021 06.
Article em En | MEDLINE | ID: mdl-34081701
ABSTRACT
Homozygous mutation of the Csf1r locus (Csf1rko) in mice, rats and humans leads to multiple postnatal developmental abnormalities. To enable analysis of the mechanisms underlying the phenotypic impacts of Csf1r mutation, we bred a rat Csf1rko allele to the inbred dark agouti (DA) genetic background and to a Csf1r-mApple reporter transgene. The Csf1rko led to almost complete loss of embryonic macrophages and ablation of most adult tissue macrophage populations. We extended previous analysis of the Csf1rko phenotype to early postnatal development to reveal impacts on musculoskeletal development and proliferation and morphogenesis in multiple organs. Expression profiling of 3-week old wild-type (WT) and Csf1rko livers identified 2760 differentially expressed genes associated with the loss of macrophages, severe hypoplasia, delayed hepatocyte maturation, disrupted lipid metabolism and the IGF1/IGF binding protein system. Older Csf1rko rats developed severe hepatic steatosis. Consistent with the developmental delay in the liver Csf1rko rats had greatly-reduced circulating IGF1. Transfer of WT bone marrow (BM) cells at weaning without conditioning repopulated resident macrophages in all organs, including microglia in the brain, and reversed the mutant phenotypes enabling long term survival and fertility. WT BM transfer restored osteoclasts, eliminated osteopetrosis, restored bone marrow cellularity and architecture and reversed granulocytosis and B cell deficiency. Csf1rko rats had an elevated circulating CSF1 concentration which was rapidly reduced to WT levels following BM transfer. However, CD43hi non-classical monocytes, absent in the Csf1rko, were not rescued and bone marrow progenitors remained unresponsive to CSF1. The results demonstrate that the Csf1rko phenotype is autonomous to BM-derived cells and indicate that BM contains a progenitor of tissue macrophages distinct from hematopoietic stem cells. The model provides a unique system in which to define the pathways of development of resident tissue macrophages and their local and systemic roles in growth and organ maturation.
Assuntos

Texto completo: 1 Bases de dados: MEDLINE Assunto principal: Osteopetrose / Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos / Desenvolvimento Musculoesquelético / Fígado Gorduroso / Macrófagos / Anormalidades Musculoesqueléticas Tipo de estudo: Prognostic_studies Idioma: En Revista: PLoS Genet Assunto da revista: GENETICA Ano de publicação: 2021 Tipo de documento: Article País de afiliação: Austrália

Texto completo: 1 Bases de dados: MEDLINE Assunto principal: Osteopetrose / Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos / Desenvolvimento Musculoesquelético / Fígado Gorduroso / Macrófagos / Anormalidades Musculoesqueléticas Tipo de estudo: Prognostic_studies Idioma: En Revista: PLoS Genet Assunto da revista: GENETICA Ano de publicação: 2021 Tipo de documento: Article País de afiliação: Austrália