Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Methods Mol Biol ; 2713: 199-206, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-37639125

RESUMO

Testis-resident macrophages are first responders of the innate immune system against pathogens. They also exert day-to-day functions that are poorly understood. To study testis macrophages, several techniques are used, among which we can find flow cytometry.Flow cytometry is a powerful tool that enables analysis of macrophages at a cellular as well as population level. To analyze testis macrophages using flow cytometry, a specific tissue processing is necessary to extract them. In this protocol, we explain how to extract and analyze the distinct macrophage populations.


Assuntos
Macrófagos , Testículo , Masculino , Humanos , Citometria de Fluxo
2.
Sci Immunol ; 8(89): eadd4374, 2023 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-37922341

RESUMO

The salivary glands often become damaged in individuals receiving radiotherapy for head and neck cancer, resulting in chronic dry mouth. This leads to detrimental effects on their health and quality of life, for which there is no regenerative therapy. Macrophages are the predominant immune cell in the salivary glands and are attractive therapeutic targets due to their unrivaled capacity to drive tissue repair. Yet, the nature and role of macrophages in salivary gland homeostasis and how they may contribute to tissue repair after injury are not well understood. Here, we show that at least two phenotypically and transcriptionally distinct CX3CR1+ macrophage populations are present in the adult salivary gland, which occupy anatomically distinct niches. CD11c+CD206-CD163- macrophages typically associate with gland epithelium, whereas CD11c-CD206+CD163+ macrophages associate with blood vessels and nerves. Using a suite of complementary fate mapping systems, we show that there are highly dynamic changes in the ontogeny and composition of salivary gland macrophages with age. Using an in vivo model of radiation-induced salivary gland injury combined with genetic or antibody-mediated depletion of macrophages, we demonstrate an essential role for macrophages in clearance of cells with DNA damage. Furthermore, we show that epithelial-associated macrophages are indispensable for effective tissue repair and gland function after radiation-induced injury, with their depletion resulting in reduced saliva production. Our data, therefore, provide a strong case for exploring the therapeutic potential of manipulating macrophages to promote tissue repair and thus minimize salivary gland dysfunction after radiotherapy.


Assuntos
Neoplasias de Cabeça e Pescoço , Xerostomia , Humanos , Macrófagos , Qualidade de Vida , Glândulas Salivares , Xerostomia/terapia
4.
Immunity ; 56(8): 1778-1793.e10, 2023 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-37463581

RESUMO

Unlike macrophage networks composed of long-lived tissue-resident cells within specific niches, conventional dendritic cells (cDCs) that generate a 3D network in lymph nodes (LNs) are short lived and continuously replaced by DC precursors (preDCs) from the bone marrow (BM). Here, we examined whether specific anatomical niches exist within which preDCs differentiate toward immature cDCs. In situ photoconversion and Prtn3-based fate-tracking revealed that the LN medullary cords are preferential entry sites for preDCs, serving as specific differentiation niches. Repopulation and fate-tracking approaches demonstrated that the cDC1 network unfolded from the medulla along the vascular tree toward the paracortex. During inflammation, collective maturation and migration of resident cDC1s to the paracortex created discontinuity in the medullary cDC1 network and temporarily impaired responsiveness. The decrease in local cDC1 density resulted in higher Flt3L availability in the medullary niche, which accelerated cDC1 development to restore the network. Thus, the spatiotemporal development of the cDC1 network is locally regulated in dedicated LN niches via sensing of cDC1 densities.


Assuntos
Linfonodos , Macrófagos , Diferenciação Celular , Células Dendríticas
5.
J Exp Med ; 220(4)2023 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-36917028

RESUMO

Tingible body macrophages in lymph node are involved in cleaning up debris from apoptotic B cells. Gurwisz et al. (2023. J. Exp. Med.https://doi.org/10.1084/jem.20222173) and Grootveld et al. (2023. Cell.https://doi.org/10.1016/j.cell.2023.02.004) report how tingible body macrophages, originating from tissue-resident macrophages, clear apoptotic B cells in the germinal center using a "stand-hunting" strategy.


Assuntos
Centro Germinativo , Linfonodos , Macrófagos , Linfócitos B , Dendritos
6.
Immunol Rev ; 315(1): 31-53, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36752151

RESUMO

Mast cells (MCs) are evolutionarily ancient innate immune cells with important roles in protective immunity against bacteria, parasites, and venomous animals. They can be found in most organs of the body, where they also contribute to normal tissue functioning, for example by engaging in crosstalk with nerves. Despite this, they are most widely known for their detrimental roles in allergy, anaphylaxis, and atopic disease. Just like macrophages, mast cells were conventionally thought to originate from the bone marrow. However, they are already present in fetal tissues before the onset of bone marrow hematopoiesis, questioning this dogma. In recent years, our view of myeloid cell ontogeny has been revised. We now know that the first mast cells originate from progenitors made in the extra-embryonic yolk sac, and later get supplemented with mast cells produced from subsequent waves of hematopoiesis. In most connective tissues, sizeable populations of fetal-derived mast cells persist into adulthood, where they self-maintain largely independently from the bone marrow. These developmental origins are highly reminiscent of macrophages, which are known to have critical functions in development. Mast cells too may thus support healthy development. Their fetal origins and longevity also make mast cells susceptible to genetic and environmental perturbations, which may render them pathological. Here, we review our current understanding of mast cell biology from a developmental perspective. We first summarize how mast cell populations are established from distinct hematopoietic progenitor waves, and how they are subsequently maintained throughout life. We then discuss what functions mast cells may normally have at early life stages, and how they may be co-opted to cause, worsen, or increase susceptibility to disease.


Assuntos
Células-Tronco Hematopoéticas , Mastócitos , Animais , Humanos , Macrófagos , Medula Óssea , Hematopoese/genética , Desenvolvimento Fetal , Diferenciação Celular
7.
Cell Rep ; 37(4): 109878, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34706240

RESUMO

Blood endothelial cells display remarkable plasticity depending on the demands of a malignant microenvironment. While studies in solid tumors focus on their role in metabolic adaptations, formation of high endothelial venules (HEVs) in lymph nodes extends their role to the organization of immune cell interactions. As a response to lymphoma growth, blood vessel density increases; however, the fate of HEVs remains elusive. Here, we report that lymphoma causes severe HEV regression in mouse models that phenocopies aggressive human B cell lymphomas. HEV dedifferentiation occurrs as a consequence of a disrupted lymph-carrying conduit system. Mechanosensitive fibroblastic reticular cells then deregulate CCL21 migration paths, followed by deterioration of dendritic cell proximity to HEVs. Loss of this crosstalk deprives HEVs of lymphotoxin-ß-receptor (LTßR) signaling, which is indispensable for their differentiation and lymphocyte transmigration. Collectively, this study reveals a remodeling cascade of the lymph node microenvironment that is detrimental for immune cell trafficking in lymphoma.


Assuntos
Movimento Celular , Células Endoteliais/metabolismo , Linfócitos/metabolismo , Linfoma de Células B/metabolismo , Animais , Células Endoteliais/patologia , Humanos , Células Jurkat , Linfócitos/patologia , Linfoma de Células B/patologia , Camundongos , Camundongos Transgênicos , Vênulas
8.
Immunol Rev ; 302(1): 104-125, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34028841

RESUMO

Macrophages are an integral part of all organs in the body, where they contribute to immune surveillance, protection, and tissue-specific homeostatic functions. This is facilitated by so-called niches composed of macrophages and their surrounding stroma. These niches structurally anchor macrophages and provide them with survival factors and tissue-specific signals that imprint their functional identity. In turn, macrophages ensure appropriate functioning of the niches they reside in. Macrophages thus form reciprocal, mutually beneficial circuits with their cellular niches. In this review, we explore how this concept applies to the spleen, a large secondary lymphoid organ whose primary functions are to filter the blood and regulate immunity. We first outline the splenic micro-anatomy, the different populations of splenic fibroblasts and macrophages and their respective contribution to protection of and key physiological processes occurring in the spleen. We then discuss firmly established and potential cellular circuits formed by splenic macrophages and fibroblasts, with an emphasis on the molecular cues underlying their crosstalk and their relevance to splenic functionality. Lastly, we conclude by considering how these macrophage-fibroblast circuits might be impaired by aging, and how understanding these changes might help identify novel therapeutic avenues with the potential of restoring splenic functions in the elderly.


Assuntos
Macrófagos , Baço , Idoso , Fibroblastos , Homeostase , Humanos , Contagem de Leucócitos
9.
Trends Immunol ; 42(2): 120-136, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33423933

RESUMO

The human body contains dozens of endocrine and exocrine glands, which regulate physiological processes by secreting hormones and other factors. Glands can be subdivided into contiguous tissue modules, each consisting of an interdependent network of cells that together perform particular tissue functions. Among those cells are macrophages, a diverse type of immune cells endowed with trophic functions. In this review, we discuss recent findings on how resident macrophages support tissue modules within glands via the creation of mutually beneficial cell-cell circuits. A better comprehension of gland macrophage function and local control within their niche is essential to achieve a refined understanding of gland physiology in homeostasis and disease.


Assuntos
Macrófagos , Homeostase , Humanos , Contagem de Leucócitos
10.
Sci Adv ; 6(48)2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33239294

RESUMO

Resident macrophages are abundant in the bladder, playing key roles in immunity to uropathogens. Yet, whether they are heterogeneous, where they come from, and how they respond to infection remain largely unknown. We identified two macrophage subsets in mouse bladders, MacM in muscle and MacL in the lamina propria, each with distinct protein expression and transcriptomes. Using a urinary tract infection model, we validated our transcriptomic analyses, finding that MacM macrophages phagocytosed more bacteria and polarized to an anti-inflammatory profile, whereas MacL macrophages died rapidly during infection. During resolution, monocyte-derived cells contributed to tissue-resident macrophage pools and both subsets acquired transcriptional profiles distinct from naïve macrophages. Macrophage depletion resulted in the induction of a type 1-biased immune response to a second urinary tract infection, improving bacterial clearance. Our study uncovers the biology of resident macrophages and their responses to an exceedingly common infection in a largely overlooked organ, the bladder.


Assuntos
Bexiga Urinária , Infecções Urinárias , Animais , Perfilação da Expressão Gênica , Macrófagos/metabolismo , Camundongos , Infecções Urinárias/metabolismo
11.
Immunity ; 53(4): 775-792.e9, 2020 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-33002412

RESUMO

Innate lymphoid cells (ILCs) are generated early during ontogeny and persist predominantly as tissue-resident cells. Here, we examined how ILCs are maintained and renewed within tissues. We generated a single cell atlas of lung ILC2s and found that Il18r1+ ILCs comprise circulating and tissue-resident ILC progenitors (ILCP) and effector-cells with heterogeneous expression of the transcription factors Tcf7 and Zbtb16, and CD103. Our analyses revealed a continuous differentiation trajectory from Il18r1+ ST2- ILCPs to Il18r- ST2+ ILC2s, which was experimentally validated. Upon helminth infection, recruited and BM-derived cells generated the entire spectrum of ILC2s in parabiotic and shield chimeric mice, consistent with their potential role in the renewal of tissue ILC2s. Our findings identify local ILCPs and reveal ILCP in situ differentiation and tissue adaptation as a mechanism of ILC maintenance and phenotypic diversification. Local niches, rather than progenitor origin, or the developmental window during ontogeny, may dominantly imprint ILC phenotypes in adult tissues.


Assuntos
Imunidade Inata/imunologia , Linfócitos/imunologia , Células Progenitoras Linfoides/imunologia , Animais , Diferenciação Celular/imunologia , Células Cultivadas , Feminino , Humanos , Subunidade alfa de Receptor de Interleucina-18/imunologia , Pulmão/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Proteína com Dedos de Zinco da Leucemia Promielocítica/imunologia , Transdução de Sinais/imunologia , Análise de Célula Única/métodos , Fator 1 de Transcrição de Linfócitos T/imunologia , Fatores de Transcrição/imunologia
12.
Nat Commun ; 11(1): 4816, 2020 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-32968047

RESUMO

Understanding cell types and mechanisms of dental growth is essential for reconstruction and engineering of teeth. Therefore, we investigated cellular composition of growing and non-growing mouse and human teeth. As a result, we report an unappreciated cellular complexity of the continuously-growing mouse incisor, which suggests a coherent model of cell dynamics enabling unarrested growth. This model relies on spatially-restricted stem, progenitor and differentiated populations in the epithelial and mesenchymal compartments underlying the coordinated expansion of two major branches of pulpal cells and diverse epithelial subtypes. Further comparisons of human and mouse teeth yield both parallelisms and differences in tissue heterogeneity and highlight the specifics behind growing and non-growing modes. Despite being similar at a coarse level, mouse and human teeth reveal molecular differences and species-specific cell subtypes suggesting possible evolutionary divergence. Overall, here we provide an atlas of human and mouse teeth with a focus on growth and differentiation.


Assuntos
Diferenciação Celular , Células-Tronco/citologia , Dente/citologia , Dente/crescimento & desenvolvimento , Adolescente , Adulto , Animais , Diferenciação Celular/genética , Células Epiteliais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Heterogeneidade Genética , Humanos , Incisivo/citologia , Incisivo/crescimento & desenvolvimento , Masculino , Mesoderma/citologia , Mesoderma/crescimento & desenvolvimento , Mesoderma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Dente Molar/citologia , Dente Molar/crescimento & desenvolvimento , Odontoblastos , Adulto Jovem
13.
Cell Rep ; 32(6): 108004, 2020 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-32783932

RESUMO

During embryogenesis, lymphoid tissue inducer (LTi) cells are essential for lymph node organogenesis. These cells are part of the innate lymphoid cell (ILC) family. Although their earliest embryonic hematopoietic origin is unclear, other innate immune cells have been shown to be derived from early hemogenic endothelium in the yolk sac as well as the aorta-gonad-mesonephros. A proper model to discriminate between these locations was unavailable. In this study, using a Cxcr4-CreERT2 lineage tracing model, we identify a major contribution from embryonic hemogenic endothelium, but not the yolk sac, toward LTi progenitors. Conversely, embryonic LTi cells are replaced by hematopoietic stem cell-derived cells in adults. We further show that, in the fetal liver, common lymphoid progenitors differentiate into highly dynamic alpha-lymphoid precursor cells that, at this embryonic stage, preferentially mature into LTi precursors and establish their functional LTi cell identity only after reaching the periphery.


Assuntos
Hemangioblastos/metabolismo , Hematopoese/fisiologia , Tecido Linfoide/embriologia , Receptores CXCR4/metabolismo , Animais , Desenvolvimento Embrionário/fisiologia , Hemangioblastos/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Imunidade Inata , Fígado/embriologia , Linfócitos/metabolismo , Linfócitos T Auxiliares-Indutores/metabolismo , Saco Vitelino/embriologia
14.
Immunity ; 53(1): 127-142.e7, 2020 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-32562599

RESUMO

Located within red pulp cords, splenic red pulp macrophages (RPMs) are constantly exposed to the blood flow, clearing senescent red blood cells (RBCs) and recycling iron from hemoglobin. Here, we studied the mechanisms underlying RPM homeostasis, focusing on the involvement of stromal cells as these cells perform anchoring and nurturing macrophage niche functions in lymph nodes and liver. Microscopy revealed that RPMs are embedded in a reticular meshwork of red pulp fibroblasts characterized by the expression of the transcription factor Wilms' Tumor 1 (WT1) and colony stimulating factor 1 (CSF1). Conditional deletion of Csf1 in WT1+ red pulp fibroblasts, but not white pulp fibroblasts, drastically altered the RPM network without altering circulating CSF1 levels. Upon RPM depletion, red pulp fibroblasts transiently produced the monocyte chemoattractants CCL2 and CCL7, thereby contributing to the replenishment of the RPM network. Thus, red pulp fibroblasts anchor and nurture RPM, a function likely conserved in humans.


Assuntos
Fibroblastos/metabolismo , Fator Estimulador de Colônias de Macrófagos/metabolismo , Macrófagos/imunologia , Baço/citologia , Proteínas WT1/metabolismo , Animais , Quimiocina CCL2/metabolismo , Quimiocina CCL7/metabolismo , Regulação da Expressão Gênica , Humanos , Imunidade Inata/imunologia , Ferro/metabolismo , Fator Estimulador de Colônias de Macrófagos/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/imunologia , Ratos , Transdução de Sinais/imunologia , Baço/metabolismo
15.
Immunity ; 52(3): 434-451, 2020 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-32187515

RESUMO

Self-maintaining resident macrophages populate all mammalian organs. In addition to their role as immune sentinels, macrophages also perform day-to-day functions essential to tissue homeostasis. The homeostatic functions of macrophages are regulated by so-called tissular "niches" that control the size of the macrophage population and imprint tissue-specific identity. Here, we review the mechanisms underlying self-maintenance of distinct macrophage populations and outline the organizing principles of the macrophage niche. We examine recent studies that uncovered mutually beneficial cell-cell circuits established between macrophages and their niche and propose a modular view of tissues that integrates the resident macrophage as an essential component of each individual module. Manipulating macrophage niche cells to control the function of resident macrophages in vivo might have therapeutic value in various disease settings.


Assuntos
Microambiente Celular/imunologia , Homeostase/imunologia , Macrófagos/imunologia , Especificidade de Órgãos/imunologia , Animais , Sobrevivência Celular/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Interleucinas/imunologia , Interleucinas/metabolismo , Fator Estimulador de Colônias de Macrófagos/imunologia , Fator Estimulador de Colônias de Macrófagos/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo
16.
J Exp Med ; 217(4)2020 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-31951251

RESUMO

Experimental and clinical evidence suggests that tumor-associated macrophages (TAMs) play important roles in cancer progression. Here, we have characterized the ontogeny and function of TAM subsets in a mouse model of metastatic ovarian cancer that is representative for visceral peritoneal metastasis. We show that the omentum is a critical premetastatic niche for development of invasive disease in this model and define a unique subset of CD163+ Tim4+ resident omental macrophages responsible for metastatic spread of ovarian cancer cells. Transcriptomic analysis showed that resident CD163+ Tim4+ omental macrophages were phenotypically distinct and maintained their resident identity during tumor growth. Selective depletion of CD163+ Tim4+ macrophages in omentum using genetic and pharmacological tools prevented tumor progression and metastatic spread of disease. These studies describe a specific role for tissue-resident macrophages in the invasive progression of metastatic ovarian cancer. The molecular pathways of cross-talk between tissue-resident macrophages and disseminated cancer cells may represent new targets to prevent metastasis and disease recurrence.


Assuntos
Macrófagos/metabolismo , Omento/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Neoplasias Peritoneais/metabolismo , Neoplasias Peritoneais/secundário , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/genética , Antígenos de Diferenciação Mielomonocítica/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Feminino , Perfilação da Expressão Gênica , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Transcriptoma
17.
Science ; 365(6460): 1461-1466, 2019 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-31604275

RESUMO

Tissue-resident immune cells are important for organ homeostasis and defense. The epithelium may contribute to these functions directly or by cross-talk with immune cells. We used single-cell RNA sequencing to resolve the spatiotemporal immune topology of the human kidney. We reveal anatomically defined expression patterns of immune genes within the epithelial compartment, with antimicrobial peptide transcripts evident in pelvic epithelium in the mature, but not fetal, kidney. A network of tissue-resident myeloid and lymphoid immune cells was evident in both fetal and mature kidney, with postnatal acquisition of transcriptional programs that promote infection-defense capabilities. Epithelial-immune cross-talk orchestrated localization of antibacterial macrophages and neutrophils to the regions of the kidney most susceptible to infection. Overall, our study provides a global overview of how the immune landscape of the human kidney is zonated to counter the dominant immunological challenge.


Assuntos
Rim/imunologia , Macrófagos/citologia , Neutrófilos/citologia , Adulto , Animais , Células Epiteliais/citologia , Feminino , Feto , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Rim/anatomia & histologia , Rim/citologia , Linfócitos/citologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Mieloides/citologia , RNA-Seq , Análise de Célula Única , Infecções Urinárias/imunologia
18.
Immunity ; 50(6): 1453-1466.e4, 2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31053503

RESUMO

In lymph nodes, subcapsular sinus macrophages (SSMs) form an immunological barrier that monitors lymph drained from peripheral tissues. Upon infection, SSMs activate B and natural killer T (NKT) cells while secreting inflammatory mediators. Here, we investigated the mechanisms regulating development and homeostasis of SSMs. Embryonic SSMs originated from yolk sac hematopoiesis and were replaced by a postnatal wave of bone marrow (BM)-derived monocytes that proliferated to establish the adult SSM network. The SSM network self-maintained by proliferation with minimal BM contribution. Upon pathogen-induced transient deletion, BM-derived cells contributed to restoring the SSM network. Lymphatic endothelial cells (LECs) were the main source of CSF-1 within the lymph node and conditional deletion of Csf1 in adult LECs decreased the network of SSMs and medullary sinus macrophages (MSMs). Thus, SSMs have a dual hematopoietic origin, and LECs are essential to the niche supporting these macrophages.


Assuntos
Células Endoteliais/metabolismo , Macrófagos/metabolismo , Animais , Biomarcadores , Comunicação Celular , Diferenciação Celular , Expressão Gênica , Genes Reporter , Hematopoese/genética , Hematopoese/imunologia , Homeostase , Linfonodos/citologia , Linfonodos/imunologia , Vasos Linfáticos , Fator Estimulador de Colônias de Macrófagos/metabolismo , Macrófagos/citologia , Macrófagos/imunologia , Camundongos , Monócitos/citologia , Monócitos/metabolismo , Saco Vitelino
19.
Cell Rep ; 26(12): 3257-3271.e8, 2019 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-30893599

RESUMO

In the bone marrow, CXCL12 and IL-7 are essential for B cell differentiation, whereas hematopoietic stem cell (HSC) maintenance requires SCF and CXCL12. Peri-sinusoidal stromal (PSS) cells are the main source of IL-7, but their characterization as a pro-B cell niche remains limited. Here, we characterize pro-B cell supporting stromal cells and decipher the interaction network allowing pro-B cell retention. Preferential contacts are found between pro-B cells and PSS cells, which homogeneously express HSC and B cell niche genes. Furthermore, pro-B cells are frequently located in the vicinity of HSCs in the same niche. Using an interactome bioinformatics pipeline, we identify Nidogen-1 as essential for pro-B cell retention in the peri-sinusoidal niche as confirmed in Nidogen-1-/- mice. Finally, human pro-B cells and hematopoietic progenitors are observed close to similar IL-7+ stromal cells. Thus, a multispecific niche exists in mouse and human supporting both early progenitors and committed hematopoietic lineages.


Assuntos
Células-Tronco Hematopoéticas/imunologia , Glicoproteínas de Membrana/imunologia , Células Precursoras de Linfócitos B/imunologia , Nicho de Células-Tronco/imunologia , Animais , Células-Tronco Hematopoéticas/citologia , Interleucina-7/genética , Interleucina-7/imunologia , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Células Precursoras de Linfócitos B/citologia , Células Estromais/citologia , Células Estromais/imunologia
20.
Science ; 363(6432)2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30872492

RESUMO

Macrophages are a heterogeneous cell population involved in tissue homeostasis, inflammation, and various pathologies. Although the major tissue-resident macrophage populations have been extensively studied, interstitial macrophages (IMs) residing within the tissue parenchyma remain poorly defined. Here we studied IMs from murine lung, fat, heart, and dermis. We identified two independent IM subpopulations that are conserved across tissues: Lyve1loMHCIIhiCX3CR1hi (Lyve1loMHCIIhi) and Lyve1hiMHCIIloCX3CR1lo (Lyve1hiMHCIIlo) monocyte-derived IMs, with distinct gene expression profiles, phenotypes, functions, and localizations. Using a new mouse model of inducible macrophage depletion (Slco2b1 flox/DTR), we found that the absence of Lyve1hiMHCIIlo IMs exacerbated experimental lung fibrosis. Thus, we demonstrate that two independent populations of IMs coexist across tissues and exhibit conserved niche-dependent functional programming.


Assuntos
Pulmão/imunologia , Pulmão/patologia , Macrófagos/imunologia , Animais , Antígenos Ly , Receptor 1 de Quimiocina CX3C/genética , Linhagem da Célula , Derme/imunologia , Modelos Animais de Doenças , Fibrose , Glicoproteínas/análise , Antígenos de Histocompatibilidade Classe II/genética , Proteínas de Membrana Transportadoras , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/imunologia , Miocárdio/imunologia , Transportadores de Ânions Orgânicos/genética , Análise de Sequência de RNA/métodos , Análise de Célula Única/métodos , Transcriptoma
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA