Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Am Coll Cardiol ; 82(20): 1921-1931, 2023 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-37940229

RESUMO

BACKGROUND: Despite major advances in pharmacological treatment for patients with heart failure, residual mortality remains high. This suggests that important pathways are not yet targeted by current heart failure therapies. OBJECTIVES: We sought integration of genetic, transcriptomic, and proteomic data in a large cohort of patients with heart failure to detect major pathways related to progression of heart failure leading to death. METHODS: We used machine learning methodology based on stacked generalization framework and gradient boosting algorithms, using 54 clinical phenotypes, 403 circulating plasma proteins, 36,046 transcript expression levels in whole blood, and 6 million genomic markers to model all-cause mortality in 2,516 patients with heart failure from the BIOSTAT-CHF (Systems BIOlogy Study to TAilored Treatment in Chronic Heart Failure) study. Results were validated in an independent cohort of 1,738 patients. RESULTS: The mean age of the patients was 70 years (Q1-Q3: 61-78 years), 27% were female, median N-terminal pro-B-type natriuretic peptide was 4,275 ng/L (Q1-Q3: 2,360-8,486 ng/L), and 7% had heart failure with preserved ejection fraction. During a median follow-up of 21 months, 657 (26%) of patients died. The 4 major pathways with a significant association to all-cause mortality were: 1) the PI3K/Akt pathway; 2) the MAPK pathway; 3) the Ras signaling pathway; and 4) epidermal growth factor receptor tyrosine kinase inhibitor resistance. Results were validated in an independent cohort of 1,738 patients. CONCLUSIONS: A systems biology approach integrating genomic, transcriptomic, and proteomic data identified 4 major pathways related to mortality. These pathways are related to decreased activation of the cardioprotective ERBB2 receptor, which can be modified by neuregulin.


Assuntos
Insuficiência Cardíaca , Proteômica , Humanos , Feminino , Idoso , Masculino , Biomarcadores , Multiômica , Fosfatidilinositol 3-Quinases/uso terapêutico , Insuficiência Cardíaca/tratamento farmacológico
2.
Life Sci Alliance ; 6(4)2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36717247

RESUMO

Systemic inflammation halts lymphopoiesis and prioritizes myeloid cell production. How blood cell production switches from homeostasis to emergency myelopoiesis is incompletely understood. Here, we show that lymphotoxin-ß receptor (LTßR) signaling in combination with TNF and IL-1 receptor signaling in bone marrow mesenchymal stem cells (MSCs) down-regulates Il7 expression to shut down lymphopoiesis during systemic inflammation. LTßR signaling in MSCs also promoted CCL2 production during systemic inflammation. Pharmacological or genetic blocking of LTßR signaling in MSCs partially enabled lymphopoiesis and reduced monocyte numbers in the spleen during systemic inflammation, which correlated with reduced survival during systemic bacterial and viral infections. Interestingly, lymphotoxin-α1ß2 delivered by B-lineage cells, and specifically by mature B cells, contributed to promote Il7 down-regulation and reduce MSC lymphopoietic activity. Our studies revealed an unexpected role of LTßR signaling in MSCs and identified recirculating mature B cells as an important regulator of emergency myelopoiesis.


Assuntos
Células-Tronco Mesenquimais , Mielopoese , Humanos , Interleucina-7 , Linfócitos B/metabolismo , Células-Tronco Mesenquimais/metabolismo , Inflamação/metabolismo
3.
Sci Immunol ; 7(75): eabo3170, 2022 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-36149943

RESUMO

Gain-of-function (GOF) mutations in CXCR4 cause WHIM (warts, hypogammaglobulinemia, infections, and myelokathexis) syndrome, characterized by infections, leukocyte retention in bone marrow (BM), and blood leukopenias. B lymphopenia is evident at early progenitor stages, yet why do CXCR4 GOF mutations that cause B (and T) lymphopenia remain obscure? Using a CXCR4 R334X GOF mouse model of WHIM syndrome, we showed that lymphopoiesis is reduced because of a dysregulated mesenchymal stem cell (MSC) transcriptome characterized by a switch from an adipogenic to an osteolineage-prone program with limited lymphopoietic activity. We identify lymphotoxin beta receptor (LTßR) as a critical pathway promoting interleukin-7 (IL-7) down-regulation in MSCs. Blocking LTßR or CXCR4 signaling restored IL-7 production and B cell development in WHIM mice. LTßR blocking also increased production of IL-7 and B cell activating factor (BAFF) in secondary lymphoid organs (SLOs), increasing B and T cell numbers in the periphery. These studies revealed that LTßR signaling in BM MSCs and SLO stromal cells limits the lymphocyte compartment size.


Assuntos
Síndromes de Imunodeficiência , Linfopenia , Animais , Fator Ativador de Células B , Síndromes de Imunodeficiência/complicações , Síndromes de Imunodeficiência/genética , Interleucina-7 , Receptor beta de Linfotoxina , Camundongos , Doenças da Imunodeficiência Primária , Nicho de Células-Tronco , Linfócitos T , Verrugas
4.
Nat Commun ; 13(1): 4611, 2022 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-35941168

RESUMO

Cellular competition for limiting hematopoietic factors is a physiologically regulated but poorly understood process. Here, we studied this phenomenon by hampering hematopoietic progenitor access to Leptin receptor+ mesenchymal stem/progenitor cells (MSPCs) and endothelial cells (ECs). We show that HSC numbers increase by 2-fold when multipotent and lineage-restricted progenitors fail to respond to CXCL12 produced by MSPCs and ECs. HSCs are qualitatively normal, and HSC expansion only occurs when early hematopoietic progenitors but not differentiated hematopoietic cells lack CXCR4. Furthermore, the MSPC and EC transcriptomic heterogeneity is stable, suggesting that it is impervious to major changes in hematopoietic progenitor interactions. Instead, HSC expansion correlates with increased availability of membrane-bound stem cell factor (mSCF) on MSPCs and ECs presumably due to reduced consumption by cKit-expressing hematopoietic progenitors. These studies suggest that an intricate homeostatic balance between HSCs and proximal hematopoietic progenitors is regulated by cell competition for limited amounts of mSCF.


Assuntos
Células Endoteliais , Células-Tronco Mesenquimais , Diferenciação Celular , Células-Tronco Hematopoéticas , Fator de Células-Tronco
5.
Cancers (Basel) ; 13(4)2021 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-33671266

RESUMO

We assessed the feasibility of adjuvant S-1 and oxaliplatin following neoadjuvant chemoradiotherapy (nCRT) and esophagectomy. Patients treated with nCRT (paclitaxel, carboplatin) and esophagectomy received six 21-day cycles with oxaliplatin (130 mg/m2) on day 1 and S-1 (25 mg/m2 twice daily) on days 1-14. The primary endpoint was feasibility, defined as ≥50% completing treatment. We performed exploratory propensity-score matching to compare survival, ERCC1 and Thymidylate Synthase (TS) immunohistochemistry analyses, proteomics biomarker discovery and 5-FU pharmacokinetic analyses. Forty patients were enrolled and 48% completed all adjuvant cycles. Median dose intensity was 98% for S-1 and 62% for oxaliplatin. The main reason for early discontinuation was toxicity (67%). The median recurrence-free and overall survival were 28.3 months and 40.8 months, respectively (median follow-up 29.1 months). Survival was not significantly prolonged compared to a matched cohort (p = 0.09). Patients with ERCC1 negative tumor expression had significantly better survival compared to ERCC1 positivity (p = 0.01). Our protein signature model was predictive of survival [p = 0.04; Area under the curve (AUC) 0.80]. Moreover, 5-FU pharmacokinetics significantly correlated with treatment-related toxicity. To conclude, six cycles adjuvant S-1 and oxaliplatin were not feasible in pretreated esophageal adenocarcinoma. Although the question remains whether additional treatment with chemotherapy should be provided in the adjuvant setting, subgroups such as patients with ERCC1 negativity could potentially benefit from adjuvant SOX based on our exploratory biomarker research.

6.
Front Immunol ; 11: 600127, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33324418

RESUMO

Studies over the last couple of decades have shown that hematopoietic stem cells (HSCs) are critically dependent on cytokines such as Stem Cell Factor and other signals provided by bone marrow niches comprising of mesenchymal stem and progenitor cells (MSPCs) and endothelial cells (ECs). Because of their critical roles in HSC maintenance the niches formed by MSPCs and ECs are commonly referred to as HSC niches. For the most part, the signals required for HSC maintenance act in a short-range manner, which imposes the necessity for directional and positional cues in order for HSCs to localize and be retained properly in stem cell niches. The chemokine CXCL12 and its Gαi protein coupled receptor CXCR4, besides promoting HSC quiescence directly, also play instrumental roles in enabling HSCs to access bone marrow stem cell niches. Recent studies have revealed, however, that HSC niches also provide a constellation of hematopoietic cytokines that are critical for the production of most, if not all, blood cell types. Some hematopoietic cytokines, namely IL-7 and IL-15 produced by HSC niches, are not only required for lymphopoiesis but are also essential for memory T cell maintenance. Consequently, hematopoietic progenitors and differentiated immune cells, such as memory T cell subsets, also depend on the CXCL12/CXCR4 axis for migration into bone marrow and interactions with MSPCs and ECs. Similarly, subsets of antibody-secreting plasma cells also reside in close association with CXCL12-producing MSPCs in the bone marrow and require the CXCR4/CXCL12 axis for survival and long-term maintenance. Collectively, these studies demonstrate a broad range of key physiological roles, spanning blood cell production and maintenance of immunological memory, that are orchestrated by stem cell niches through a common and simple mechanism: CXCL12/CXCR4-mediated cell recruitment followed by receipt of a maintenance and/or instructive signal. A fundamental flaw of this type of cellular organization is revealed by myeloid and lymphoid leukemias, which target stem cell niches and induce profound transcriptomic changes that result in reduced hematopoietic activity and altered mesenchymal cell differentiation.


Assuntos
Células-Tronco Hematopoéticas/imunologia , Memória Imunológica , Transdução de Sinais/imunologia , Nicho de Células-Tronco/imunologia , Animais , Células Endoteliais/imunologia , Humanos , Células-Tronco Mesenquimais/imunologia
7.
EBioMedicine ; 61: 103079, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33096472

RESUMO

BACKGROUND: Familial hypercholesterolemia (FH) is a monogenic disorder characterized by elevated low-density lipoprotein cholesterol (LDL-C). A FH causing genetic variant in LDLR, APOB, or PCSK9 is not identified in 12-60% of clinical FH patients (FH mutation-negative patients). We aimed to assess whether altered DNA methylation might be associated with FH in this latter group. METHODS: In this study we included 78 FH mutation-negative patients and 58 FH mutation-positive patients with a pathogenic LDLR variant. All patients were male, not using lipid lowering therapies and had LDL-C levels >6 mmol/L and triglyceride levels <3.5 mmol/L. DNA methylation was measured with the Infinium Methylation EPIC 850 K beadchip assay. Multiple linear regression analyses were used to explore DNA methylation differences between the two groups in genes related to lipid metabolism. A gradient boosting machine learning model was applied to investigate accumulated genome-wide differences between the two groups. FINDINGS: Candidate gene analysis revealed one significantly hypomethylated CpG site in CPT1A (cg00574958) in FH mutation-negative patients, while no differences in methylation in other lipid genes were observed. The machine learning model did distinguish the two groups with a mean Area Under the Curve (AUC)±SD of 0.80±0.17 and provided two CpG sites (cg26426080 and cg11478607) in genes with a possible link to lipid metabolism (PRDM16 and GSTT1). INTERPRETATION: FH mutation-negative patients are characterized by accumulated genome wide DNA methylation differences, but not by major DNA methylation alterations in known lipid genes compared to FH mutation-positive patients. FUNDING: ZonMW grant (VIDI no. 016.156.445).


Assuntos
Metilação de DNA , Predisposição Genética para Doença , Hiperlipoproteinemia Tipo II/etiologia , Adolescente , Adulto , Biomarcadores , Biologia Computacional/métodos , Ilhas de CpG , Epigênese Genética , Epigenômica/métodos , Regulação da Expressão Gênica , Humanos , Hiperlipoproteinemia Tipo II/diagnóstico , Hiperlipoproteinemia Tipo II/metabolismo , Aprendizado de Máquina , Pessoa de Meia-Idade , Mutação , Curva ROC , Adulto Jovem
8.
Cell Stem Cell ; 27(2): 189-190, 2020 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-32763177

RESUMO

Stem cell biologists have been yearning to visualize hematopoietic stem cells (HSCs) in live animals since Kiel et al. (2005) first visualized them in bone cavities. With two recent papers from Christodoulou et al. (2020) and Upadhaya et al. (2020), we can all now see how HSCs behave in their niches!


Assuntos
Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas , Animais
9.
Am J Dermatopathol ; 42(2): 125-128, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31268927

RESUMO

Histoid leprosy (HL) was originally described by Wade in 1963 and is regarded as a rare variant of lepromatous leprosy (LL). These characteristic clinical lesions are firm, deeply adhered nodules with features reminiscent of dermatofibromas or keloids in a background of apparently healthy skin. The main histopathological findings described are the presence of spindle cell histiocytes immersed in a richly collagenized background, usually forming a nodular pattern of infiltration with sharply delimitation and positive staining for acid-fast bacilli. The classical form of HL lesions should be devoid of foam histiocytes and globi. However, we and other authors noticed that in most of the cases, despite characteristic clinical features, histopathology depicts a mixture of LL and HL patterns. Therefore, we present a case with clinical features similar to HL in which an excisional scalpel biopsy of a nodule demonstrated features of classical LL in the center of the lesion and features of HL in the periphery, highlighting that a proper biopsy technique could enhance the ability of the dermatopathologist to histopathologically diagnose cases of HL. In cases in which HL is clinically suspected, we advocate replacing the usual 4-mm incisional punch biopsy by a broader elliptical scalpel biopsy, encompassing the totality of the lesion whenever possible to achieve a reliable representation of the pathologic process.


Assuntos
Biópsia/métodos , Hanseníase Virchowiana/diagnóstico , Hanseníase Virchowiana/patologia , Feminino , Histiócitos/patologia , Humanos , Adulto Jovem
10.
Immunol Rev ; 289(1): 142-157, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30977190

RESUMO

Studies over the last decade uncovered overlapping niches for hematopoietic stem cells (HSCs), multipotent progenitor cells, common lymphoid progenitors, and early B cell progenitors. HSC and lymphoid niches are predominantly composed by mesenchymal progenitor cells (MPCs) and by a small subset of endothelial cells. Niche cells create specialized microenvironments through the concomitant production of short-range acting cell-fate determining cytokines such as interleukin (IL)-7 and stem cell factor and the potent chemoattractant C-X-C motif chemokine ligand 12. This type of cellular organization allows for the cross-talk between hematopoietic stem and progenitor cells with niche cells, such that niche cell activity can be regulated by the quality and quantity of hematopoietic progenitors being produced. For example, preleukemic B cell progenitors and preB acute lymphoblastic leukemias interact directly with MPCs, and downregulate IL-7 expression and the production of non-leukemic lymphoid cells. In this review, we discuss a novel model of B cell development that is centered on cellular circuits formed between B cell progenitors and lymphopoietic niches.


Assuntos
Linfócitos B/fisiologia , Quimiocina CXCL12/metabolismo , Células Endoteliais/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Células-Tronco Mesenquimais/fisiologia , Animais , Diferenciação Celular , Microambiente Celular , Humanos , Interleucina-7/metabolismo , Linfopoese , Nicho de Células-Tronco
11.
J Exp Med ; 215(10): 2586-2599, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30158115

RESUMO

B cell progenitors require paracrine signals such as interleukin-7 (IL-7) provided by bone marrow stromal cells for proliferation and survival. Yet, how B cells regulate access to these signals in vivo remains unclear. Here we show that proB and IL-7+ cells form a cell circuit wired by IL-7R signaling, which controls CXCR4 and focal adhesion kinase (FAK) expression and restricts proB cell movement due to increased adhesion to IL-7+CXCL12Hi cells. PreBCR signaling breaks this circuit by switching the preB cell behavior into a fast-moving and lower-adhesion state via increased CXCR4 and reduced FAK/α4ß1 expression. This behavioral change reduces preB cell exposure to IL-7, thereby attenuating IL-7R signaling in vivo. Remarkably, IL-7 production is downregulated by signals provided by preB cells with unrepaired double-stranded DNA breaks and by preB acute lymphoblastic leukemic cells. Combined, these studies revealed that distinct cell circuits control the quality and homeostasis of B cell progenitors.


Assuntos
Interleucina-7/imunologia , Células-Tronco Mesenquimais/imunologia , Células Precursoras de Linfócitos B/imunologia , Receptores de Interleucina-7/imunologia , Transdução de Sinais/imunologia , Animais , Quimiocina CXCL12/genética , Interleucina-7/genética , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Knockout , Células Precursoras de Linfócitos B/citologia , Receptores de Interleucina-7/genética , Transdução de Sinais/genética
12.
J Immunol ; 201(3): 908-915, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29934471

RESUMO

Recirculation of naive T cells between secondary lymphoid organs to receive survival cues and scan for signs of infection or other pathologic conditions is important for immune homeostasis and effective immune responses. Although the mechanisms that specifically guide the entry of naive T cells into secondary lymphoid organs are well studied, the mechanisms that keep them from fluxing into inappropriate or undesirable compartments, such as healthy tissues or bone marrow, are less well understood. In this study, we report an unexpected finding that under steady state, bone marrow homing of naive T cells is actively suppressed by mTORC2 signaling. We found that in mice, T cell-specific deletion of an essential mTORC2 component Sin1 results in increased accumulation of naive T cells in the bone marrow. Mechanistically, we show that loss of mTORC2 signaling in naive T cells results in enhanced FOXO1 activity, which leads to increased CXCR4 expression and chemotactic response to CXCL12, a key chemokine that promotes bone marrow homing and retention of T cells. Together, the results of our study reveal a novel role of mTORC2 in T cell homeostasis via active suppression of naive T cell bone marrow homing by the mTORC2-FOXO1-CXCR4 axis.


Assuntos
Medula Óssea/imunologia , Medula Óssea/metabolismo , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Receptores CXCR4/metabolismo , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Animais , Proteínas de Transporte/metabolismo , Quimiocina CXCL12/metabolismo , Proteína Forkhead Box O1/metabolismo , Homeostase/imunologia , Camundongos , Camundongos Endogâmicos C57BL
13.
Adv Immunol ; 134: 47-88, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28413023

RESUMO

B lymphocytes develop from hematopoietic stem cells (HSCs) in specialized bone marrow niches composed of rare mesenchymal lineage stem/progenitor cells (MSPCs) and sinusoidal endothelial cells. These niches are defined by function and location: MSPCs are mostly perisinusoidal cells that together with a small subset of sinusoidal endothelial cells express stem cell factor, interleukin-7 (IL-7), IL-15, and the highest amounts of CXCL12 in bone marrow. Though rare, MSPCs are morphologically heterogeneous, highly reticular, and form a vast cellular network in the bone marrow parenchyma capable of interacting with large numbers of hematopoietic cells. HSCs, downstream multipotent progenitor cells, and common lymphoid progenitor cells utilize CXCR4 to fine-tune access to critical short-range growth factors provided by MSPCs for their long-term maintenance and/or multilineage differentiation. In later stages, developing B lymphocytes use CXCR4 to navigate the bone marrow parenchyma, and predominantly cannabinoid receptor-2 for positioning within bone marrow sinusoids, prior to being released into peripheral blood circulation. In the final stages of differentiation, transitional B cells migrate to the spleen where they preferentially undergo further rounds of differentiation until selection into the mature B cell pool occurs. This bottleneck purges up to 97% of all developing B cells in a peripheral selection process that is heavily controlled not only by the intensity of BCR signaling and access to BAFF but also by the proper functioning of the B cell motility machinery.


Assuntos
Linfócitos B/citologia , Diferenciação Celular/imunologia , Quimiocinas/imunologia , Células-Tronco Hematopoéticas/citologia , Linfopoese/imunologia , Animais , Linfócitos B/imunologia , Células-Tronco Hematopoéticas/imunologia , Humanos
14.
Clin Rev Allergy Immunol ; 51(1): 59-78, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26511861

RESUMO

Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease that primarily affects the joints. Self-reactive B and T lymphocytes cooperate to promote antibody responses against self proteins and are major drivers of disease. T lymphocytes also promote RA independently of B lymphocytes mainly through the production of key inflammatory cytokines, such as IL-17, that promote pathology. While the innate signals that initiate self-reactive adaptive immune responses are poorly understood, the disease is predominantly caused by inflammatory cellular infiltration and accumulation in articular tissues, and by bone erosions driven by bone-resorbing osteoclasts. Osteoclasts are giant multinucleated cells formed by the fusion of multiple myeloid cells that require short-range signals, such as the cytokines MCSF and RANKL, for undergoing differentiation. The recruitment and positioning of osteoclast precursors to sites of osteoclast differentiation by chemoattractants is an important point of control for osteoclastogenesis and bone resorption. Recently, the GPCR EBI2 and its oxysterol ligand 7a, 25 dihydroxycholesterol, were identified as important regulators of osteoclast precursor positioning in proximity to bone surfaces and of osteoclast differentiation under homeostasis. In chronic inflammatory diseases like RA, osteoclast differentiation is also driven by inflammatory cytokines such as TNFa and IL-1, and can occur independently of RANKL. Finally, there is growing evidence that the chemotactic signals guiding osteoclast precursors to inflamed articular sites contribute to disease and are of great interest. Furthering our understanding of the complex osteoimmune cell interactions should provide new avenues of therapeutic intervention for RA.


Assuntos
Artrite Reumatoide/etiologia , Artrite Reumatoide/metabolismo , Movimento Celular , Osteoclastos/metabolismo , Animais , Artrite Reumatoide/patologia , Medula Óssea/imunologia , Medula Óssea/metabolismo , Medula Óssea/patologia , Diferenciação Celular , Microambiente Celular , Quimiotaxia , Homeostase , Humanos , Imunidade Inata , Linfócitos/imunologia , Linfócitos/metabolismo , Osteoclastos/citologia , Ligante RANK/metabolismo , Membrana Sinovial/imunologia , Membrana Sinovial/metabolismo , Membrana Sinovial/patologia
15.
J Exp Med ; 212(11): 1931-46, 2015 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-26438360

RESUMO

Bone surfaces attract hematopoietic and nonhematopoietic cells, such as osteoclasts (OCs) and osteoblasts (OBs), and are targeted by bone metastatic cancers. However, the mechanisms guiding cells toward bone surfaces are essentially unknown. Here, we show that the Gαi protein-coupled receptor (GPCR) EBI2 is expressed in mouse monocyte/OC precursors (OCPs) and its oxysterol ligand 7α,25-dihydroxycholesterol (7α,25-OHC) is secreted abundantly by OBs. Using in vitro time-lapse microscopy and intravital two-photon microscopy, we show that EBI2 enhances the development of large OCs by promoting OCP motility, thus facilitating cell-cell interactions and fusion in vitro and in vivo. EBI2 is also necessary and sufficient for guiding OCPs toward bone surfaces. Interestingly, OCPs also secrete 7α,25-OHC, which promotes autocrine EBI2 signaling and reduces OCP migration toward bone surfaces in vivo. Defective EBI2 signaling led to increased bone mass in male mice and protected female mice from age- and estrogen deficiency-induced osteoporosis. This study identifies a novel pathway involved in OCP homing to the bone surface that may have significant therapeutic potential.


Assuntos
Densidade Óssea/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Hidroxicolesteróis/farmacologia , Osteoclastos/efeitos dos fármacos , Receptores Acoplados a Proteínas G/fisiologia , Células-Tronco/efeitos dos fármacos , Animais , Osso e Ossos/citologia , Diferenciação Celular/efeitos dos fármacos , Feminino , Homeostase , Camundongos , Camundongos Endogâmicos C57BL , Osteoclastos/fisiologia , Transdução de Sinais , Células-Tronco/fisiologia
16.
PLoS One ; 10(8): e0136061, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26274929

RESUMO

Suppressor of cytokine signaling (SOCS)-3 has been suggested to regulate CXCR4 signaling in a variety of human cell lines. In mice, conditional SOCS3 inactivation in hematopoietic cells including B-lineage lymphocytes has been reported to exacerbate CXCR4-signaling and focal adhesion kinase phosphorylation, which resulted in altered immature B cell distribution in bone marrow (BM) due to sustained α4ß1 integrin-mediated adhesion to the extracellular matrix. However, a recent study examining conditional SOCS3 deletion specifically in B-lineage cells failed to detect significant roles in B-lineage cell retention in BM. In this study we carefully examined the role played by SOCS3 in CXCR4 signaling in developing B cell subsets. We show that in mice conditionally deficient in SOCS3 exclusively in B cells (Socs3fl/fl Mb1cre/+) there was no detectable difference in B cell development in BM and in periphery. We show that SOCS3 deficient and sufficient immature B cell subsets are similarly distributed between BM parenchyma and sinusoids, and are equally competent at exiting BM into peripheral blood. Furthermore, we found no significant differences in CXCR4 desensitization upon ligand exposure in developing B lymphocyte subsets. Consequently, SOCS3-deficient and sufficient B-lineage cell migration towards CXCL12 in vitro was undistinguishable, and B-lineage cell amoeboid motility within BM parenchyma was also unaffected by SOCS3-deficiency. Thus we conclude that SOCS3 has no detectable influence on biological processes known to be controlled by CXCR4 signaling.


Assuntos
Subpopulações de Linfócitos B/metabolismo , Medula Óssea/metabolismo , Movimento Celular/fisiologia , Transdução de Sinais/fisiologia , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Animais , Subpopulações de Linfócitos B/citologia , Adesão Celular/fisiologia , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Humanos , Integrina alfa4/genética , Integrina alfa4/metabolismo , Cadeias beta de Integrinas/genética , Cadeias beta de Integrinas/metabolismo , Camundongos , Camundongos Transgênicos , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/genética
17.
J Exp Med ; 211(13): 2567-81, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25403444

RESUMO

Leukocyte residence in lymphoid organs is controlled by a balance between retention and egress-promoting chemoattractants sensed by pertussis toxin (PTX)-sensitive Gαi protein-coupled receptors (GPCRs). Here, we use two-photon intravital microscopy to show that immature B cell retention within bone marrow (BM) was strictly dependent on amoeboid motility mediated by CXCR4 and CXCL12 and by α4ß1 integrin-mediated adhesion to VCAM-1. However, B lineage cell egress from BM is independent of PTX-sensitive GPCR signaling. B lineage cells expressing PTX rapidly exited BM even though their motility within BM parenchyma was significantly reduced. Our experiments reveal that when immature B cells are near BM sinusoids their motility is reduced, their morphology is predominantly rounded, and cells reverse transmigrate across sinusoidal endothelium in a largely nonamoeboid manner. Immature B cell egress from BM was dependent on a twofold CXCR4 down-regulation that was antagonized by antigen-induced BCR signaling. This passive mode of cell egress from BM also contributes significantly to the export of other hematopoietic cells, including granulocytes, monocytes, and NK cells, and is reminiscent of erythrocyte egress.


Assuntos
Linfócitos B/citologia , Células da Medula Óssea/citologia , Diferenciação Celular/imunologia , Receptores CXCR4/metabolismo , Animais , Linhagem da Célula , Movimento Celular , Forma Celular , Regulação para Baixo , Integrina alfa4beta1/metabolismo , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos B/metabolismo , Receptores CXCR4/antagonistas & inibidores , Transdução de Sinais , Molécula 1 de Adesão de Célula Vascular/metabolismo
18.
J Exp Med ; 206(11): 2469-81, 2009 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-19808259

RESUMO

During a screen for ethylnitrosourea-induced mutations in mice affecting blood natural killer (NK) cells, we identified a strain, designated Duane, in which NK cells were reduced in blood and spleen but increased in lymph nodes (LNs) and bone marrow (BM). The accumulation of NK cells in LNs reflected a decreased ability to exit into lymph. This strain carries a point mutation within Tbx21 (T-bet), which generates a defective protein. Duane NK cells have a 30-fold deficiency in sphingosine-1-phosphate receptor 5 (S1P5) transcript levels, and S1P5-deficient mice exhibit an egress defect similar to Duane. Chromatin immunoprecipitation confirms binding of T-bet to the S1pr5 locus. S1P-deficient mice exhibit a more severe NK cell egress block, and the FTY720-sensitive S1P1 also plays a role in NK cell egress from LNs. S1P5 is not inhibited by CD69, a property that may facilitate trafficking of activated NK cells to effector sites. Finally, the accumulation of NK cells within BM of S1P-deficient mice was associated with reduced numbers in BM sinusoids, suggesting a role for S1P in BM egress. In summary, these findings identify S1P5 as a T-bet-induced gene that is required for NK cell egress from LNs and BM.


Assuntos
Células da Medula Óssea/citologia , Movimento Celular , Células Matadoras Naturais/citologia , Células Matadoras Naturais/metabolismo , Linfonodos/citologia , Receptores de Lisoesfingolipídeo/metabolismo , Proteínas com Domínio T/metabolismo , Animais , Células da Medula Óssea/efeitos dos fármacos , Contagem de Células , Movimento Celular/efeitos dos fármacos , Etilnitrosoureia , Cloridrato de Fingolimode , Células Matadoras Naturais/efeitos dos fármacos , Linfonodos/efeitos dos fármacos , Ativação Linfocitária/efeitos dos fármacos , Lisofosfolipídeos/deficiência , Lisofosfolipídeos/farmacologia , Camundongos , Camundongos Mutantes , Mutação/genética , Propilenoglicóis/farmacologia , Esfingosina/análogos & derivados , Esfingosina/deficiência , Esfingosina/farmacologia
19.
Nat Immunol ; 10(4): 403-11, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19252491

RESUMO

Immature B cells developing in the bone marrow are found in the parenchyma and sinusoids. The mechanisms that control the positioning of B cells in the sinusoids are not understood. Here we show that the integrin alpha(4)beta(1) (VLA-4) and its ligand VCAM-1 were required, whereas the chemokine receptor CXCR4 was dispensable, for sinusoidal retention of B cells. Instead, cannabinoid receptor 2 (CB2), a Galpha(i) protein-coupled receptor upregulated in immature B cells, was required for sinusoidal retention. Using two-photon microscopy, we found immature B cells entering and crawling in sinusoids; these immature B cells were displaced by CB2 antagonism. Moreover, CB2-deficient mice had a lower frequency of immunoglobulin lambda-chain-positive B cells in the peripheral blood and spleen. Our findings identify unique requirements for the retention of B cells in the bone marrow sinusoidal niche and suggest involvement of CB2 in the generation of the B cell repertoire.


Assuntos
Medula Óssea/fisiologia , Células Precursoras de Linfócitos B/fisiologia , Receptor CB2 de Canabinoide/fisiologia , Animais , Medula Óssea/imunologia , Movimento Celular/fisiologia , Subunidades alfa de Proteínas de Ligação ao GTP/fisiologia , Cadeias lambda de Imunoglobulina/sangue , Cadeias lambda de Imunoglobulina/imunologia , Integrina alfa4beta1/imunologia , Integrina alfa4beta1/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Precursoras de Linfócitos B/citologia , Células Precursoras de Linfócitos B/imunologia , Receptor CB2 de Canabinoide/genética , Receptor CB2 de Canabinoide/imunologia , Receptores CXCR4/imunologia , Receptores CXCR4/metabolismo , Baço/imunologia , Regulação para Cima , Molécula 1 de Adesão de Célula Vascular/imunologia , Molécula 1 de Adesão de Célula Vascular/fisiologia
20.
Science ; 316(5822): 295-8, 2007 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-17363629

RESUMO

Lymphocytes require sphingosine-1-phosphate (S1P) receptor-1 to exit lymphoid organs, but the source(s) of extracellular S1P and whether S1P directly promotes egress are unknown. By using mice in which the two kinases that generate S1P were conditionally ablated, we find that plasma S1P is mainly hematopoietic in origin, with erythrocytes a major contributor, whereas lymph S1P is from a distinct radiation-resistant source. Lymphocyte egress from thymus and secondary lymphoid organs was markedly reduced in kinase-deficient mice. Restoration of S1P to plasma rescued egress to blood but not lymph, and the rescue required lymphocyte expression of S1P-receptor-1. Thus, separate sources provide S1P to plasma and lymph to help lymphocytes exit the low-S1P environment of lymphoid organs. Disruption of compartmentalized S1P signaling is a plausible mechanism by which S1P-receptor-1 agonists function as immunosuppressives.


Assuntos
Medula Óssea/metabolismo , Linfócitos/fisiologia , Lisofosfolipídeos/biossíntese , Lisofosfolipídeos/fisiologia , Esfingosina/análogos & derivados , Animais , Quimiotaxia de Leucócito/fisiologia , Cromatografia Líquida , Endotélio Vascular , Feminino , Células-Tronco Hematopoéticas/metabolismo , Linfócitos/metabolismo , Lisofosfolipídeos/sangue , Lisofosfolipídeos/deficiência , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Receptores de Lisoesfingolipídeo/fisiologia , Esfingosina/biossíntese , Esfingosina/sangue , Esfingosina/deficiência , Esfingosina/fisiologia , Espectrometria de Massas em Tandem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA