Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
Add more filters

Publication year range
1.
EMBO J ; 2024 May 30.
Article in English | MEDLINE | ID: mdl-38816652

ABSTRACT

In mice, γδ-T lymphocytes that express the co-stimulatory molecule, CD27, are committed to the IFNγ-producing lineage during thymic development. In the periphery, these cells play a critical role in host defense and anti-tumor immunity. Unlike αß-T cells that rely on MHC-presented peptides to drive their terminal differentiation, it is unclear whether MHC-unrestricted γδ-T cells undergo further functional maturation after exiting the thymus. Here, we provide evidence of phenotypic and functional diversity within peripheral IFNγ-producing γδ T cells. We found that CD27+ Ly6C- cells convert into CD27+Ly6C+ cells, and these CD27+Ly6C+ cells control cancer progression in mice, while the CD27+Ly6C- cells cannot. The gene signatures of these two subsets were highly analogous to human immature and mature γδ-T cells, indicative of conservation across species. We show that IL-27 supports the cytotoxic phenotype and function of mouse CD27+Ly6C+ cells and human Vδ2+ cells, while IL-27 is dispensable for mouse CD27+Ly6C- cell and human Vδ1+ cell functions. These data reveal increased complexity within IFNγ-producing γδ-T cells, comprising immature and terminally differentiated subsets, that offer new insights into unconventional T-cell biology.

2.
Immunity ; 50(2): 378-389.e5, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30784579

ABSTRACT

Currently, we lack an understanding of the individual and combinatorial roles for chemokine receptors in the inflammatory process. We report studies on mice with a compound deletion of Ccr1, Ccr2, Ccr3, and Ccr5, which together control monocytic and eosinophilic recruitment to resting and inflamed sites. Analysis of resting tissues from these mice, and mice deficient in each individual receptor, provides clear evidence for redundant use of these receptors in establishing tissue-resident monocytic cell populations. In contrast, analysis of cellular recruitment to inflamed sites provides evidence of specificity of receptor use for distinct leukocyte subtypes and no indication of comprehensive redundancy. We find no evidence of involvement of any of these receptors in the recruitment of neutrophils or lymphocytes to resting or acutely inflamed tissues. Our data shed important light on combinatorial inflammatory chemokine receptor function and highlight Ccr2 as the primary driver of myelomonocytic cell recruitment in acutely inflamed contexts.


Subject(s)
Eosinophils/immunology , Inflammation/immunology , Monocytes/immunology , Receptors, CCR/immunology , Animals , Chemokines/immunology , Chemokines/metabolism , Eosinophils/metabolism , Gene Expression Profiling/methods , Inflammation/genetics , Inflammation/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Receptors, CCR/genetics , Receptors, CCR/metabolism , Receptors, CCR1/immunology , Receptors, CCR1/metabolism , Receptors, CCR2/immunology , Receptors, CCR2/metabolism , Receptors, CCR3/immunology , Receptors, CCR3/metabolism , Receptors, CCR5/immunology , Receptors, CCR5/metabolism
3.
Development ; 151(4)2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38300826

ABSTRACT

ACKR3 scavenges and degrades the stem cell recruiting chemokine CXCL12, which is essential for proper embryonic and, in particular, haematopoietic development. Here, we demonstrate strong expression of ACKR3 on trophoblasts. Using a maternally administered pharmacological blocker and Cre-mediated genetic approaches, we demonstrate that trophoblast ACKR3 is essential for preventing movement of CXCL12 from the mother to the embryo, with elevated plasma CXCL12 levels being detected in embryos from ACKR3-blocker-treated mothers. Mice born to mothers treated with the blocker are lighter and shorter than those born to vehicle-treated mothers and, in addition, display profound anaemia associated with a markedly reduced bone marrow haematopoietic stem cell population. Importantly, although the haematopoietic abnormalities are corrected as mice age, our studies reveal a postnatal window during which offspring of ACKR3-blocker-treated mice are unable to mount effective inflammatory responses to inflammatory/infectious stimuli. Overall, these data demonstrate that ACKR3 is essential for preventing CXCL12 transfer from mother to embryo and for ensuring properly regulated CXCL12 control over the development of the haematopoietic system.


Subject(s)
Placenta , Receptors, CXCR , Animals , Female , Mice , Pregnancy , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Movement , Mutation , Placenta/metabolism , Receptors, CXCR/genetics , Receptors, CXCR/metabolism , Signal Transduction/genetics
4.
J Immunol ; 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38829123

ABSTRACT

The interactions between chemokines and their receptors, particularly in the context of inflammation, are complex, with individual receptors binding multiple ligands and individual ligands interacting with multiple receptors. In addition, there are numerous reports of simultaneous coexpression of multiple inflammatory chemokine receptors on individual inflammatory leukocyte subtypes. Overall, this has previously been interpreted as redundancy and proposed as a protective mechanism to ensure that the inflammatory response is robust. By contrast, we have hypothesized that the system is not redundant but exquisitely subtle. Our interests relate to the receptors CCR1, CCR2, CCR3, and CCR5, which, together, regulate nonneutrophilic myeloid cell recruitment to inflammatory sites. In this study, we demonstrate that although most murine monocytes exclusively express CCR2, there is a small subpopulation that is expanded during inflammation and coexpresses CCR1 and CCR2. Combinations of transcript and functional analysis demonstrate that this is not redundant expression and that coexpression of CCR1 and CCR2 marks a phenotypically distinct population of monocytes characterized by expression of genes otherwise typically associated with neutrophils. Single-cell RNA sequencing confirms this as a monodisperse population of atypical monocytes. This monocytic population has previously been described as having immunosuppressive activity. Overall, our data confirm combinatorial chemokine receptor expression by a subpopulation of monocytes but demonstrate that this is not redundant expression and marks a discrete monocytic population.

5.
Immunity ; 44(6): 1455-69, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27332734

ABSTRACT

Aedes aegypti mosquitoes are responsible for transmitting many medically important viruses such as those that cause Zika and dengue. The inoculation of viruses into mosquito bite sites is an important and common stage of all mosquito-borne virus infections. We show, using Semliki Forest virus and Bunyamwera virus, that these viruses use this inflammatory niche to aid their replication and dissemination in vivo. Mosquito bites were characterized by an edema that retained virus at the inoculation site and an inflammatory influx of neutrophils that coordinated a localized innate immune program that inadvertently facilitated virus infection by encouraging the entry and infection of virus-permissive myeloid cells. Neutrophil depletion and therapeutic blockade of inflammasome activity suppressed inflammation and abrogated the ability of the bite to promote infection. This study identifies facets of mosquito bite inflammation that are important determinants of the subsequent systemic course and clinical outcome of virus infection.


Subject(s)
Arbovirus Infections/immunology , Bunyamwera virus/physiology , Inflammation/immunology , Insect Bites and Stings/immunology , Neutrophils/immunology , Semliki forest virus/physiology , Virus Replication , Animals , Cell Movement , Cells, Cultured , Culicidae/immunology , Humans , Immunity, Innate , Inflammasomes/metabolism , Inflammation/virology , Insect Bites and Stings/virology , Mice , Neutrophils/virology
6.
Circulation ; 147(12): 956-972, 2023 03 21.
Article in English | MEDLINE | ID: mdl-36484244

ABSTRACT

BACKGROUND: Placental heart development and embryonic heart development occur in parallel, and these organs have been proposed to exert reciprocal regulation during gestation. Poor placentation has been associated with congenital heart disease, an important cause of infant mortality. However, the mechanisms by which altered placental development can lead to congenital heart disease remain unresolved. METHODS: In this study, we use an in vivo neutrophil-driven placental inflammation model through antibody depletion of maternal circulating neutrophils at key stages during time-mated murine pregnancy: embryonic days 4.5 and 7.5. Pregnant mice were culled at embryonic day 14.5 to assess placental and embryonic heart development. A combination of flow cytometry, histology, and bulk RNA sequencing was used to assess placental immune cell composition and tissue architecture. We also used flow cytometry and single-cell sequencing to assess embryonic cardiac immune cells at embryonic day 14.5 and histology and gene analyses to investigate embryonic heart structure and development. In some cases, offspring were culled at postnatal days 5 and 28 to assess any postnatal cardiac changes in immune cells, structure, and cardiac function, as measured by echocardiography. RESULTS: In the present study, we show that neutrophil-driven placental inflammation leads to inadequate placental development and loss of barrier function. Consequently, placental inflammatory monocytes of maternal origin become capable of migration to the embryonic heart and alter the normal composition of resident cardiac macrophages and cardiac tissue structure. This cardiac impairment continues into postnatal life, hindering normal tissue architecture and function. Last, we show that tempering placental inflammation can prevent this fetal cardiac defect and is sufficient to promote normal cardiac function in postnatal life. CONCLUSIONS: Taken together, these observations provide a mechanistic paradigm whereby neutrophil-driven inflammation in pregnancy can preclude normal embryonic heart development as a direct consequence of poor placental development, which has major implications on cardiac function into adult life.


Subject(s)
Heart Defects, Congenital , Placenta , Pregnancy , Female , Mice , Animals , Placenta/pathology , Placentation , Fetus , Inflammation/pathology
7.
Development ; 147(12)2020 06 17.
Article in English | MEDLINE | ID: mdl-32467242

ABSTRACT

Macrophages are key regulators of developmental processes, including those involved in mammary gland development. We have previously demonstrated that the atypical chemokine receptor ACKR2 contributes to the control of ductal epithelial branching in the developing mammary gland by regulating macrophage dynamics. ACKR2 is a chemokine-scavenging receptor that mediates its effects through collaboration with inflammatory chemokine receptors (iCCRs). Here, we reveal reciprocal regulation of branching morphogenesis in the mammary gland, whereby stromal ACKR2 modulates levels of the shared ligand CCL7 to control the movement of a key population of CCR1-expressing macrophages to the ductal epithelium. In addition, oestrogen, which is essential for ductal elongation during puberty, upregulates CCR1 expression on macrophages. The age at which girls develop breasts is decreasing, which raises the risk of diseases including breast cancer. This study presents a previously unknown mechanism controlling the rate of mammary gland development during puberty and highlights potential therapeutic targets.


Subject(s)
Macrophages/metabolism , Mammary Glands, Animal/growth & development , Receptors, Chemokine/metabolism , Animals , Chemokine CCL3/deficiency , Chemokine CCL3/genetics , Chemokine CCL3/metabolism , Chemokine CCL5/deficiency , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Epithelium/metabolism , Estradiol/pharmacology , Female , Lectins, C-Type/metabolism , Macrophages/cytology , Mammary Glands, Animal/metabolism , Mannose Receptor , Mannose-Binding Lectins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis , Receptors, CCR1/deficiency , Receptors, CCR1/genetics , Receptors, CCR1/metabolism , Receptors, Cell Surface/metabolism , Receptors, Chemokine/deficiency , Receptors, Chemokine/genetics , Up-Regulation/drug effects
8.
Int J Mol Sci ; 24(12)2023 Jun 14.
Article in English | MEDLINE | ID: mdl-37373278

ABSTRACT

Mesenchymal stromal cells (MSC) show promise as cellular therapeutics. Psoriasis is a chronic inflammatory disease affecting the skin and the joints. Injury, trauma, infection and medications can trigger psoriasis by disrupting epidermal keratinocyte proliferation and differentiation, which activates the innate immune system. Pro-inflammatory cytokine secretion drives a T helper 17 response and an imbalance of regulatory T cells. We hypothesized that MSC adoptive cellular therapy could immunomodulate and suppress the effector T cell hyperactivation that underlies the disease. We used the imiquimod-induced psoriasis-like skin inflammation model to study the therapeutic potential of bone marrow and adipose tissue-derived MSC in vivo. We compared the secretome and the in vivo therapeutic potential of MSC with and without cytokine pre-challenge ("licensing"). The infusion of both unlicensed and licensed MSC accelerated the healing of psoriatic lesions, and reduced epidermal thickness and CD3+ T cell infiltration while promoting the upregulation of IL-17A and TGF-ß. Concomitantly, the expression of keratinocyte differentiation markers in the skin was decreased. However, unlicensed MSC promoted the resolution of skin inflammation more efficiently. We show that MSC adoptive therapy upregulates the transcription and secretion of pro-regenerative and immunomodulatory molecules in the psoriatic lesion. Accelerated healing is associated with the secretion of TGF-ß and IL-6 in the skin and MSC drives the production of IL-17A and restrains T-cell-mediated pathology.


Subject(s)
Dermatitis , Mesenchymal Stem Cells , Psoriasis , Animals , Mice , Interleukin-6/metabolism , Transforming Growth Factor beta/metabolism , Interleukin-17/metabolism , Psoriasis/drug therapy , Skin/metabolism , Cytokines/metabolism , Dermatitis/metabolism , Inflammation/metabolism , Mesenchymal Stem Cells/metabolism
9.
Gut ; 2022 Apr 27.
Article in English | MEDLINE | ID: mdl-35477863

ABSTRACT

OBJECTIVE: Hepatocellular carcinoma (HCC) is increasingly associated with non-alcoholic steatohepatitis (NASH). HCC immunotherapy offers great promise; however, recent data suggests NASH-HCC may be less sensitive to conventional immune checkpoint inhibition (ICI). We hypothesised that targeting neutrophils using a CXCR2 small molecule inhibitor may sensitise NASH-HCC to ICI therapy. DESIGN: Neutrophil infiltration was characterised in human HCC and mouse models of HCC. Late-stage intervention with anti-PD1 and/or a CXCR2 inhibitor was performed in murine models of NASH-HCC. The tumour immune microenvironment was characterised by imaging mass cytometry, RNA-seq and flow cytometry. RESULTS: Neutrophils expressing CXCR2, a receptor crucial to neutrophil recruitment in acute-injury, are highly represented in human NASH-HCC. In models of NASH-HCC lacking response to ICI, the combination of a CXCR2 antagonist with anti-PD1 suppressed tumour burden and extended survival. Combination therapy increased intratumoural XCR1+ dendritic cell activation and CD8+ T cell numbers which are associated with anti-tumoural immunity, this was confirmed by loss of therapeutic effect on genetic impairment of myeloid cell recruitment, neutralisation of the XCR1-ligand XCL1 or depletion of CD8+ T cells. Therapeutic benefit was accompanied by an unexpected increase in tumour-associated neutrophils (TANs) which switched from a protumour to anti-tumour progenitor-like neutrophil phenotype. Reprogrammed TANs were found in direct contact with CD8+ T cells in clusters that were enriched for the cytotoxic anti-tumoural protease granzyme B. Neutrophil reprogramming was not observed in the circulation indicative of the combination therapy selectively influencing TANs. CONCLUSION: CXCR2-inhibition induces reprogramming of the tumour immune microenvironment that promotes ICI in NASH-HCC.

10.
Immunology ; 165(2): 206-218, 2022 02.
Article in English | MEDLINE | ID: mdl-34775606

ABSTRACT

The immune system plays fundamental roles in the mammary gland, shaping developmental processes and controlling inflammation during infection and cancer.Here, we reveal unanticipated heterogeneity in the myeloid cell compartment duringdevelopment of virgin, pregnant, lactating and involuting mouse mammary glands,and in milk. We investigate the functional consequences of individual and compoundchemokine receptor deficiency on cell recruitment. Diverse myeloid cell recruitmentwas also shown in models of sterile inflammation and bacterial infection.Strikingly, we have shown that inflammation and infection can alter the abundanceof terminal end buds, a key developmental structure, within the pubertal mammarygland. This previously unknown effect of inflammatory burden during puberty couldhave important implications for understanding pubertal development.


Subject(s)
Disease Susceptibility , Mastitis/etiology , Mastitis/metabolism , Myeloid Cells/immunology , Myeloid Cells/metabolism , Animals , Biomarkers , Biopsy , Cellular Microenvironment/genetics , Cellular Microenvironment/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Flow Cytometry , Granulocytes/immunology , Granulocytes/metabolism , Immunohistochemistry , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Mammary Glands, Animal/immunology , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mastitis/pathology , Mice , Monocytes/immunology , Monocytes/metabolism , Myeloid Cells/pathology
11.
Trends Immunol ; 40(6): 472-481, 2019 06.
Article in English | MEDLINE | ID: mdl-31006548

ABSTRACT

Recruitment of immune cells from the vasculature relies on the presentation of glycosaminoglycan-bound chemokines on the luminal side of vascular endothelial cells. However, the current model of chemokine-glycosaminoglycan interactions, and its implications for receptor interactions, remains poorly developed. We propose a refined 'Chemokine Cloud' model, arguing that chemokines are not presented to leukocytes bound to glycosaminoglycans, but rather, in solution while sequestered within the hydrated glycocalyx. We posit that glycosaminoglycans provide an immobilized chemokine depot maintaining a 'cloud' of 'solution-phase' chemokines within the glycocalyx, and that it is this soluble form of any given chemokine that interacts with leukocyte-bound receptors. Our proposition clarifies certain anomalies associated with the current model of chemokine-glycosaminoglycan interactions, with implications for the design of blockers of chemokine function.


Subject(s)
Cell Adhesion , Leukocytes/immunology , Leukocytes/metabolism , Animals , Cell Adhesion/physiology , Chemokines/chemistry , Chemokines/metabolism , Endothelial Cells/metabolism , Glycosaminoglycans/metabolism , Humans , Leukocytes/pathology , Models, Biological , Protein Binding , Protein Multimerization , Receptors, Chemokine/metabolism , Signal Transduction
12.
PLoS Biol ; 17(5): e3000287, 2019 05.
Article in English | MEDLINE | ID: mdl-31141500

ABSTRACT

Atypical chemokine receptor 2 (ACKR2) is a chemokine-scavenging receptor. ACKR2-/-embryos display a reduction in size of a novel, to our knowledge, embryonic skin macrophage population referred to as 'intermediate' cells. CC chemokine receptor 2 (CCR2)-/-embryos display an identical phenotype, indicating that these cells require CCR2 to enable them to populate embryonic skin. Further analysis revealed that ACKR2-/-embryos have higher circulating concentrations of the CCR2 ligand, CC ligand 2 (CCL2); thus, ACKR2 regulates intraembryonic CCL2 levels. We show that ACKR2 is strongly expressed by trophoblasts and that it blocks movement of inflammatory chemokines, such as CCL2, from the maternal decidua into the embryonic circulation. We propose that trophoblastic ACKR2 is responsible for ensuring chemokine compartmentalisation on the maternal decidua, without which chemokines enter the embryonic circulation, disrupting gradients essential for directed intraembryonic cell migration. Overall, therefore, we describe a novel, to our knowledge, molecular mechanism whereby maternal decidual chemokines can function in a compartmentalised fashion without interfering with intraembryonic leukocyte migration. These data suggest similar functions for other atypical chemokine receptors in the placenta and indicate that defects in such receptors may have unanticipated developmental consequences.


Subject(s)
Chemokines/metabolism , Mammals/metabolism , Placenta/metabolism , Animals , Cell Movement , Decidua/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , Macrophages/metabolism , Mice, Inbred C57BL , Monocytes/metabolism , Pregnancy , Receptors, Chemokine/deficiency , Receptors, Chemokine/metabolism , Skin/embryology , Skin/metabolism , Transcription, Genetic , Yolk Sac/metabolism
13.
Eur J Immunol ; 50(5): 666-675, 2020 05.
Article in English | MEDLINE | ID: mdl-32114694

ABSTRACT

Analysis of chemokine receptor, and atypical chemokine receptor, expression is frequently hampered by the lack of availability of high-quality antibodies and the species specificity of those that are available. We have previously described methodology utilizing Alexa-Fluor-labeled chemokine ligands as versatile reagents to detect receptor expression. Previously this has been limited to hematopoietic cells and methodology for assessing expression of receptors on stromal cells has been lacking. Among chemokine receptors, the ones most frequently expressed on stromal cells belong to the atypical chemokine receptor subfamily. These receptors do not signal in the classic sense in response to ligand but scavenge their ligands and degrade them and thus sculpt in vivo chemokine gradients. Here, we demonstrate the ability to use either intratracheal or intravenous, Alexa-Fluor-labeled chemokine administration to detect stromal cell populations expressing the atypical chemokine receptor ACKR2. Using this methodology, we demonstrate, for the first time, expression of ACKR2 on blood endothelial cells. This observation sets the lung aside from other tissues in which ACKR2 is exclusively expressed on lymphatic endothelial cells and suggest unique roles for ACKR2 in the pulmonary environment.


Subject(s)
Endothelial Cells/immunology , Lung/immunology , Receptors, Chemokine/immunology , Stromal Cells/immunology , Animals , Carbocyanines/chemistry , Endothelial Cells/cytology , Epithelial Cell Adhesion Molecule/genetics , Epithelial Cell Adhesion Molecule/immunology , Flow Cytometry , Fluorescent Dyes/chemistry , Gene Expression , Lung/blood supply , Lung/cytology , Mice , Mice, Knockout , Receptors, Chemokine/genetics , Staining and Labeling/methods , Stromal Cells/cytology
14.
J Transl Med ; 19(1): 156, 2021 04 17.
Article in English | MEDLINE | ID: mdl-33865426

ABSTRACT

Multipotent mesenchymal stromal cells (MSCs) are promising cellular therapeutics for the treatment of inflammatory and degenerative disorders due to their anti-inflammatory, immunomodulatory and regenerative potentials. MSCs can be sourced from a variety of tissues within the body, but bone marrow is the most frequently used starting material for clinical use. The chemokine family contains many regulators of inflammation, cellular function and cellular migration-all critical factors in understanding the potential potency of a novel cellular therapeutic. In this review, we focus on expression of chemokine receptors and chemokine ligands by MSCs isolated from different tissues. We discuss the differential migratory, angiogenetic and immunomodulatory potential to understand the role that tissue source of MSC may play within a clinical context. Furthermore, this is strongly associated with leukocyte recruitment, immunomodulatory potential and T cell inhibition potential and we hypothesize that chemokine profiling can be used to predict the in vivo therapeutic potential of MSCs isolated from new sources and compare them to BM MSCs.


Subject(s)
Chemokines , Mesenchymal Stem Cells , Receptors, Chemokine , Bone Marrow Cells , Cell Differentiation , Cell Proliferation , Cells, Cultured , Humans , Immunomodulation
16.
Nat Immunol ; 10(1): 101-8, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19060902

ABSTRACT

The Duffy antigen receptor for chemokines (DARC) belongs to a family of 'silent' heptahelical chemokine receptors that do not couple to G proteins and fail to transmit measurable intracellular signals. DARC binds most inflammatory chemokines and is prominently expressed on venular endothelial cells, where its function has remained contentious. Here we show that DARC, like other silent receptors, internalized chemokines but did not effectively scavenge them. Instead, DARC mediated chemokine transcytosis, which led to apical retention of intact chemokines and more leukocyte migration across monolayers expressing DARC. Mice overexpressing DARC on blood vessel endothelium had enhanced chemokine-induced leukocyte extravasation and contact-hypersensitivity reactions. Thus, interactions of chemokines with DARC support their activity on apposing leukocytes in vitro and in vivo.


Subject(s)
Cell Movement , Chemokines/metabolism , Duffy Blood-Group System/metabolism , Leukocytes/immunology , Receptors, Cell Surface/metabolism , Animals , Cells, Cultured , Dogs , Duffy Blood-Group System/genetics , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Humans , Mice , Protein Transport , Receptors, Cell Surface/genetics
17.
Rheumatology (Oxford) ; 60(7): 3307-3316, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33313931

ABSTRACT

OBJECTIVES: Pregnancy in SSc is burdened with an increased risk of obstetric complications. Little is known about the underlying placental alterations. This study aimed to better understand pathological changes and the role of inflammation in SSc placentas. Leucocyte infiltration, inflammatory mediators and atypical chemokine receptor 2 (ACKR2) expression in SSc placentas were compared with those in other rheumatic diseases (ORD) and healthy controls (HC). METHODS: A case-control study was conducted on eight pregnant SSc patients compared with 16 patients with ORD and 16 HC matched for gestational age. Clinical data were collected. Placentas were obtained for histopathological analysis and immunohistochemistry (CD3, CD20, CD11c, CD68, ACKR2). Samples from four SSc, eight ORD and eight HC were analysed by qPCR for ACKR2 expression and by multiplex assay for cytokines, chemokines and growth factors involved in angiogenesis and inflammation. RESULTS: The number of placental CD3, CD68 and CD11 cells was significantly higher in patients affected by rheumatic diseases (SSc+ORD) compared with HC. Hepatocyte growth factor was significantly increased in the group of rheumatic diseases patients (SSc+ORD) compared with HC, while chemokine (C-C motif) ligand 5 (CCL5) was significantly higher in SSc patients compared with ORD and HC. CCL5 levels directly correlated with the number of all local inflammatory cells and higher levels were associated with histological villitis. CONCLUSIONS: Inflammatory alterations characterize placentas from rheumatic disease patients and could predispose to obstetric complications in these subjects.


Subject(s)
Cytokines/metabolism , Leukocytes/metabolism , Placenta/metabolism , Scleroderma, Systemic/metabolism , Adult , Antigens, CD/metabolism , Antigens, CD20/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Arthritis, Juvenile/metabolism , CD11c Antigen/metabolism , CD3 Complex/metabolism , Case-Control Studies , Chemokine CCL5/metabolism , Female , Fetal Membranes, Premature Rupture/metabolism , HELLP Syndrome/metabolism , Hepatocyte Growth Factor/metabolism , Humans , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/metabolism , Leukocytes/pathology , Lupus Erythematosus, Systemic/metabolism , Placenta/pathology , Pre-Eclampsia/metabolism , Pregnancy , Premature Birth/metabolism , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Rheumatic Diseases/metabolism , Sjogren's Syndrome/metabolism , Undifferentiated Connective Tissue Diseases/metabolism
18.
Development ; 144(1): 74-82, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27888192

ABSTRACT

Macrophages are important regulators of branching morphogenesis during development and postnatally in the mammary gland. Regulation of macrophage dynamics during these processes can therefore have a profound impact on development. We demonstrate here that the developing mammary gland expresses high levels of inflammatory CC-chemokines, which are essential in vivo regulators of macrophage migration. We further demonstrate that the atypical chemokine receptor ACKR2, which scavenges inflammatory CC-chemokines, is differentially expressed during mammary gland development. We have previously shown that ACKR2 regulates macrophage dynamics during lymphatic vessel development. Here, we extend these observations to reveal a novel role for ACKR2 in regulating the postnatal development of the mammary gland. Specifically, we show that Ackr2-/- mice display precocious mammary gland development. This is associated with increased macrophage recruitment to the developing gland and increased density of the ductal epithelial network. These data demonstrate that ACKR2 is an important regulator of branching morphogenesis in diverse biological contexts and provide the first evidence of a role for chemokines and their receptors in postnatal development processes.


Subject(s)
Mammary Glands, Animal/embryology , Morphogenesis/genetics , Receptors, CCR/physiology , Receptors, Chemokine/physiology , Animals , Cell Movement/genetics , Embryo, Mammalian , Female , Lymphangiogenesis/genetics , Lymphatic Vessels/embryology , Lymphatic Vessels/physiology , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Stromal Cells/metabolism
19.
J Neuroinflammation ; 17(1): 94, 2020 Mar 25.
Article in English | MEDLINE | ID: mdl-32213184

ABSTRACT

BACKGROUND: Recent years have seen an explosion of research pertaining to biological psychiatry, yet despite subsequent advances in our understanding of neuroimmune communication pathways, how the brain senses and responds to peripheral inflammation remains poorly understood. A better understanding of these pathways may be important for generating novel therapeutics to treat many patients with chronic inflammatory diseases who also suffer from neuropsychiatric comorbidities. Here we have systematically assessed the leukocyte infiltrate to the brain following systemic endotoxin exposure to better understand this novel route of neuroimmune communication. METHODS: Mice were injected intraperitoneally with LPS daily for 2, 5 or 7 consecutive days. We systematically interrogated the subsequent induction of chemokine transcription in the brain using TaqMan low-density arrays. A combination of flow cytometry and immunohistochemistry was then used to characterise the accompanying leukocyte infiltrate. RESULTS: Repeated LPS challenges resulted in prolonged activation of brain-resident microglia, coupled with an increased local transcription of numerous chemokines. After 2 days of administering LPS, there was a marked increase in the expression of the neutrophil chemoattractants CXCL1 and CXCL2; the monocyte chemoattractants CCL2, CCL5, CCL7 and CCL8; and the lymphocyte chemoattractants CXCL9, CXCL10 and CXCL16. In a number of cases, this response was sustained for several days. Chemokine induction was associated with a transient recruitment of neutrophils and monocytes to the brain, coupled with a sustained accumulation of macrophages, CD8+ T cells, NK cells and NKT cells. Strikingly, neutrophils, monocytes and T cells appeared to extravasate from the vasculature and/or CSF to infiltrate the brain parenchyma. CONCLUSIONS: Prolonged exposure to a peripheral inflammatory stimulus triggers the recruitment of myeloid cells and lymphocytes to the brain. By altering the inflammatory or metabolic milieu of the brain, this novel method of immune-to-brain communication may have profound implications for patients with chronic inflammatory diseases, potentially leading to neuropsychiatric comorbidities.


Subject(s)
Brain/drug effects , Chemokines/metabolism , Endotoxins/pharmacology , Leukocytes/drug effects , Lipopolysaccharides/pharmacology , Animals , Brain/metabolism , Inflammation/metabolism , Leukocytes/metabolism , Mice , Neutrophil Infiltration/physiology
20.
Cytotherapy ; 22(12): 762-771, 2020 12.
Article in English | MEDLINE | ID: mdl-32828673

ABSTRACT

BACKGROUND AIMS: Mesenchymal stromal cells (MSCs) isolated from various tissues are under investigation as cellular therapeutics in a wide range of diseases. It is appreciated that the basic biological functions of MSCs vary depending on tissue source. However, in-depth comparative analyses between MSCs isolated from different tissue sources under Good Manufacturing Practice (GMP) conditions are lacking. Human clinical-grade low-purity islet (LPI) fractions are generated as a byproduct of islet isolation for transplantation. MSC isolates were derived from LPI fractions with the aim of performing a systematic, standardized comparative analysis of these cells with clinically relevant bone marrow-derived MSCs (BM MSCs). METHODS: MSC isolates were derived from LPI fractions and expanded in platelet lysate-supplemented medium or in commercially available xenogeneic-free medium. Doubling rate, phenotype, differentiation potential, gene expression, protein production and immunomodulatory capacity of LPIs were compared with those of BM MSCs. RESULTS: MSCs can be readily derived in vitro from non-transplanted fractions resulting from islet cell processing (i.e., LPI MSCs). LPI MSCs grow stably in serum-free or platelet lysate-supplemented media and demonstrate in vitro self-renewal, as measured by colony-forming unit assay. LPI MSCs express patterns of chemokines and pro-regenerative factors similar to those of BM MSCs and, importantly, are equally able to attract immune cells in vitro and in vivo and suppress T-cell proliferation in vitro. Additionally, LPI MSCs can be expanded to therapeutically relevant doses at low passage under GMP conditions. CONCLUSIONS: LPI MSCs represent an alternative source of GMP MSCs with functions comparable to BM MSCs.


Subject(s)
Bone Marrow Cells/cytology , Cell Culture Techniques/methods , Immunity , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/immunology , Neovascularization, Physiologic , Pancreas/cytology , Biomarkers/metabolism , Cell Differentiation , Cell Proliferation , Cell Shape , Cells, Cultured , Colony-Forming Units Assay , Humans , Immunomodulation , Interferon-gamma/metabolism , Regenerative Medicine , T-Lymphocytes/cytology
SELECTION OF CITATIONS
SEARCH DETAIL