Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Clin Transl Sci ; 16(10): 1856-1865, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37547990

RESUMO

Pharmacological challenge models are deployed to evaluate drug effects during clinical development. Intradermal injection of Substance P (SP) neuropeptide, a potential challenge agent for investigating local mediators, is associated with wheal and flare response mediated by the MRGPRX2 receptor. Although dose-dependent data on SP effects exist, full characterization and information on potential carryover effect after repeated challenge are lacking. This open-label, two-part, prospective enabling study of SP intradermal challenge in healthy participants aimed to understand and distinguish between wheal and flare responses following various SP doses. Part 1 included one challenge visit to determine optimum SP dose range for evaluation in part 2, which determined variability in 20 participants and used intradermal microdialysis (IDM) for SP-challenged skin sampling. At 5, 15, 50, and 150 pmol doses, respectively, posterior median area under the curve (AUC; AUC0-2h ) was 4090.4, 5881.2, 8846.8, and 9212.8 mm2 /min, for wheal response, and 12020.9, 38154.3, 65470.6, and 67404.4 mm2 /min for flare response (SP-challenge visit 2). When the challenge was repeated ~2 weeks later, no carryover effect was observed. IDM histamine levels were relatively low, resulting in low confidence in the data to define temporal characteristics for histamine release following SP challenge. No safety concerns were identified using SP. Wheal and flare responses following intradermal SP challenge were dose-dependent and different. The results indicate that this challenge model is fit-for-purpose in future first-in-human studies and further assessment of novel drugs targeting dermal inflammatory disease responses, such as chronic spontaneous urticaria, chronic inducible urticaria, and pseudo-allergic reactions.


Assuntos
Hipersensibilidade , Substância P , Humanos , Histamina/sangue , Proteínas do Tecido Nervoso , Estudos Prospectivos , Receptores Acoplados a Proteínas G , Receptores de Neuropeptídeos , Pele , Substância P/farmacologia
2.
Exp Dermatol ; 30(11): 1686-1692, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33336508

RESUMO

The immunoregulator spleen tyrosine kinase (SYK) is upregulated in cutaneous lupus erythematosus (CLE). This double-blind, multicentre, Phase Ib study evaluated the safety, tolerability, pharmacokinetics, pharmacodynamics and clinical efficacy of the selective SYK inhibitor GSK2646264 in active CLE lesions. Two lesions from each participant (n = 11) were each randomized to topical application of 1% (w/w) GSK2646264 or placebo for 28 days; all participants received GSK2646264 and placebo. The primary endpoint was safety and tolerability of GSK2646264, assessed by adverse event incidence and a skin tolerability test. Secondary endpoints included change from baseline in clinical activity and mRNA expression of interferon-related genes in skin biopsies. Levels of several immune cell markers were evaluated over time. Eight (73%) participants experienced ≥ 1 adverse event (all mild in intensity), and maximal dermal response was similar for GSK2646264 and placebo. The expression of several interferon-related genes, including CXCL10 and OAS1, showed modest decreases from baseline after 28 days of treatment with GSK2646264 compared with placebo. Similar findings were observed for CD3 + T cell and CD11c + dendritic cell levels; however, overall clinical activity remained unchanged with GSK2646264 vs. placebo. Further studies are warranted to assess SYK inhibitors as potential treatment for CLE.


Assuntos
Lúpus Eritematoso Cutâneo/tratamento farmacológico , Piridinas/farmacologia , Piridinas/uso terapêutico , Quinase Syk/antagonistas & inibidores , Administração Tópica , Adulto , Idoso , Método Duplo-Cego , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Piridinas/administração & dosagem , Piridinas/farmacocinética , Resultado do Tratamento
3.
Lancet ; 391(10140): 2619-2630, 2018 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-29910042

RESUMO

BACKGROUND: B cells produce alloantibodies and activate alloreactive T cells, negatively affecting kidney transplant survival. By contrast, regulatory B cells are associated with transplant tolerance. Immunotherapies are needed that inhibit B-cell effector function, including antibody secretion, while sparing regulators and minimising infection risk. B lymphocyte stimulator (BLyS) is a cytokine that promotes B-cell activation and has not previously been targeted in kidney transplant recipients. We aimed to determine the safety and activity of an anti-BLyS antibody, belimumab, in addition to standard-of-care immunosuppression in adult kidney transplant recipients. We used an experimental medicine study design with multiple secondary and exploratory endpoints to gain further insight into the effect of belimumab on the generation of de-novo IgG and on the regulatory B-cell compartment. METHODS: We undertook a double-blind, randomised, placebo-controlled phase 2 trial of belimumab, in addition to standard-of-care immunosuppression (basiliximab, mycophenolate mofetil, tacrolimus, and prednisolone) at two centres, Addenbrooke's Hospital, Cambridge, UK, and Guy's and St Thomas' Hospital, London, UK. Participants were eligible if they were aged 18-75 years and receiving a kidney transplant and were planned to receive standard-of-care immunosuppression. Participants were randomly assigned (1:1) to receive either intravenous belimumab 10 mg per kg bodyweight or placebo, given at day 0, 14, and 28, and then every 4 weeks for a total of seven infusions. The co-primary endpoints were safety and change in the concentration of naive B cells from baseline to week 24, both of which were analysed in all patients who received a transplant and at least one dose of drug or placebo (the modified intention-to-treat [mITT] population). This trial has been completed and is registered with ClinicalTrials.gov, NCT01536379, and EudraCT, 2011-006215-56. FINDINGS: Between Sept 13, 2013, and Feb 8, 2015, of 303 patients assessed for eligibility, 28 kidney transplant recipients were randomly assigned to receive belimumab (n=14) or placebo (n=14). 25 patients (12 [86%] patients assigned to the belimumab group and 13 [93%] patients assigned to the placebo group) received a transplant and were included in the mITT population. We observed similar proportions of adverse events in the belimumab and placebo groups, including serious infections (one [8%] of 12 in the belimumab group and five [38%] of 13 in the placebo group during the 6-month on-treatment phase; and none in the belimumab group and two [15%] in the placebo group during the 6-month follow-up). In the on-treatment phase, one patient in the placebo group died because of fatal myocardial infarction and acute cardiac failure. The co-primary endpoint of a reduction in naive B cells from baseline to week 24 was not met. Treatment with belimumab did not significantly reduce the number of naive B cells from baseline to week 24 (adjusted mean difference between the belimumab and placebo treatment groups -34·4 cells per µL, 95% CI -109·5 to 40·7). INTERPRETATION: Belimumab might be a useful adjunct to standard-of-care immunosuppression in renal transplantation, with no major increased risk of infection and potential beneficial effects on humoral alloimmunity. FUNDING: GlaxoSmithKline.


Assuntos
Anticorpos Monoclonais Humanizados/administração & dosagem , Sobrevivência de Enxerto/efeitos dos fármacos , Terapia de Imunossupressão/métodos , Imunossupressores/administração & dosagem , Transplante de Rim/métodos , Administração Intravenosa , Adulto , Idoso , Método Duplo-Cego , Feminino , Humanos , Imunoglobulina G/sangue , Masculino , Pessoa de Meia-Idade
4.
Cancer Cell ; 32(3): 324-341.e6, 2017 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-28870739

RESUMO

The biological and clinical behaviors of hematological malignancies can be influenced by the active crosstalk with an altered bone marrow (BM) microenvironment. In the present study, we provide a detailed picture of the BM vasculature in acute myeloid leukemia using intravital two-photon microscopy. We found several abnormalities in the vascular architecture and function in patient-derived xenografts (PDX), such as vascular leakiness and increased hypoxia. Transcriptomic analysis in endothelial cells identified nitric oxide (NO) as major mediator of this phenotype in PDX and in patient-derived biopsies. Moreover, induction chemotherapy failing to restore normal vasculature was associated with a poor prognosis. Inhibition of NO production reduced vascular permeability, preserved normal hematopoietic stem cell function, and improved treatment response in PDX.


Assuntos
Antineoplásicos/uso terapêutico , Medula Óssea/patologia , Permeabilidade Capilar , Microambiente Celular , Progressão da Doença , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/patologia , Animais , Antineoplásicos/farmacologia , Medula Óssea/efeitos dos fármacos , Permeabilidade Capilar/efeitos dos fármacos , Microambiente Celular/efeitos dos fármacos , Perfilação da Expressão Gênica , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Humanos , Leucemia Mieloide Aguda/genética , Camundongos , Transplante de Neoplasias/patologia , Óxido Nítrico/metabolismo , Resultado do Tratamento
5.
J Vis Exp ; (126)2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28809828

RESUMO

Human hematopoietic stem cells (HSCs) reside in the bone marrow (BM) niche, an intricate, multifactorial network of components producing cytokines, growth factors, and extracellular matrix. The ability of HSCs to remain quiescent, self-renew or differentiate, and acquire mutations and become malignant depends upon the complex interactions they establish with different stromal components. To observe the crosstalk between human HSCs and the human BM niche in physiological and pathological conditions, we designed a protocol to ectopically model and image a humanized BM niche in immunodeficient mice. We show that the use of different cellular components allows for the formation of humanized structures and the opportunity to sustain long-term human hematopoietic engraftment. Using two-photon microscopy, we can live-image these structures in situ at the single-cell resolution, providing a powerful new tool for the functional characterization of the human BM microenvironment and its role in regulating normal and malignant hematopoiesis.


Assuntos
Bioengenharia/métodos , Medula Óssea/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Imagem Óptica/métodos , Alicerces Teciduais , Animais , Células da Medula Óssea/metabolismo , Proteína Morfogenética Óssea 2/metabolismo , Colágeno/química , Feminino , Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/citologia , Humanos , Masculino , Camundongos SCID , Microscopia de Fluorescência por Excitação Multifotônica/instrumentação , Microscopia de Fluorescência por Excitação Multifotônica/métodos , Imagem Óptica/instrumentação , Nicho de Células-Tronco/fisiologia
6.
J Clin Invest ; 127(2): 543-548, 2017 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-28067666

RESUMO

The BM niche comprises a tightly controlled microenvironment formed by specific tissue and cells that regulates the behavior of hematopoietic stem cells (HSCs). Here, we have provided a 3D model that is tunable in different BM niche components and useful, both in vitro and in vivo, for studying the maintenance of normal and malignant hematopoiesis. Using scaffolds, we tested the capacity of different stromal cell types to support human HSCs. Scaffolds coated with human mesenchymal stromal cells (hMSCs) proved to be superior in terms of HSC engraftment and long-term maintenance when implanted in vivo. Moreover, we found that hMSC-coated scaffolds can be modulated to form humanized bone tissue, which was also able to support human HSC engraftment. Importantly, hMSC-coated humanized scaffolds were able to support the growth of leukemia patient cells in vivo, including the growth of samples that would not engraft the BM of immunodeficient mice. These results demonstrate that an s.c. implantation approach in a 3D carrier scaffold seeded with stromal cells is an effective in vivo niche model for studying human hematopoiesis. The various niche components of this model can be changed depending on the context to improve the engraftment of nonengrafting acute myeloid leukemia (AML) samples.


Assuntos
Hematopoese/imunologia , Células-Tronco Hematopoéticas/imunologia , Leucemia Mieloide Aguda/imunologia , Células-Tronco Mesenquimais/imunologia , Modelos Biológicos , Nicho de Células-Tronco/imunologia , Microambiente Tumoral/imunologia , Animais , Células-Tronco Hematopoéticas/patologia , Humanos , Leucemia Mieloide Aguda/patologia , Células-Tronco Mesenquimais/patologia , Camundongos
7.
Blood ; 129(8): 950-958, 2017 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-27899358

RESUMO

Adhesion is a key component of hematopoietic stem cell regulation mediating homing and retention to the niche in the bone marrow. Here, using an RNA interference screen, we identify cytohesin 1 (CYTH1) as a critical mediator of adhesive properties in primary human cord blood-derived hematopoietic stem and progenitor cells (HSPCs). Knockdown of CYTH1 disrupted adhesion of HSPCs to primary human mesenchymal stroma cells. Attachment to fibronectin and ICAM1, 2 integrin ligands, was severely impaired, and CYTH1-deficient cells showed a reduced integrin ß1 activation response, suggesting that CYTH1 mediates integrin-dependent functions. Transplantation of CYTH1-knockdown cells to immunodeficient mice resulted in significantly lower long-term engraftment levels, associated with a reduced capacity of the transplanted cells to home to the bone marrow. Intravital microscopy showed that CYTH1 deficiency profoundly affects HSPC mobility and localization within the marrow space and thereby impairs proper lodgment into the niche. Thus, CYTH1 is a novel major regulator of adhesion and engraftment in human HSPCs through mechanisms that, at least in part, involve the activation of integrins.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/metabolismo , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Animais , Antígenos CD/metabolismo , Antígenos CD34/metabolismo , Adesão Celular , Moléculas de Adesão Celular/metabolismo , Movimento Celular , Fibronectinas/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Células-Tronco Hematopoéticas/metabolismo , Humanos , Integrinas/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos Endogâmicos , Interferência de RNA
8.
Nature ; 538(7626): 518-522, 2016 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-27750279

RESUMO

It is widely accepted that complex interactions between cancer cells and their surrounding microenvironment contribute to disease development, chemo-resistance and disease relapse. In light of this observed interdependency, novel therapeutic interventions that target specific cancer stroma cell lineages and their interactions are being sought. Here we studied a mouse model of human T-cell acute lymphoblastic leukaemia (T-ALL) and used intravital microscopy to monitor the progression of disease within the bone marrow at both the tissue-wide and single-cell level over time, from bone marrow seeding to development/selection of chemo-resistance. We observed highly dynamic cellular interactions and promiscuous distribution of leukaemia cells that migrated across the bone marrow, without showing any preferential association with bone marrow sub-compartments. Unexpectedly, this behaviour was maintained throughout disease development, from the earliest bone marrow seeding to response and resistance to chemotherapy. Our results reveal that T-ALL cells do not depend on specific bone marrow microenvironments for propagation of disease, nor for the selection of chemo-resistant clones, suggesting that a stochastic mechanism underlies these processes. Yet, although T-ALL infiltration and progression are independent of the stroma, accumulated disease burden leads to rapid, selective remodelling of the endosteal space, resulting in a complete loss of mature osteoblastic cells while perivascular cells are maintained. This outcome leads to a shift in the balance of endogenous bone marrow stroma, towards a composition associated with less efficient haematopoietic stem cell function. This novel, dynamic analysis of T-ALL interactions with the bone marrow microenvironment in vivo, supported by evidence from human T-ALL samples, highlights that future therapeutic interventions should target the migration and promiscuous interactions of cancer cells with the surrounding microenvironment, rather than specific bone marrow stroma, to combat the invasion by and survival of chemo-resistant T-ALL cells.


Assuntos
Células da Medula Óssea/citologia , Leucemia-Linfoma de Células T do Adulto/patologia , Transplante de Neoplasias , Microambiente Tumoral , Animais , Movimento Celular , Progressão da Doença , Feminino , Células-Tronco Hematopoéticas/citologia , Humanos , Microscopia Intravital , Masculino , Camundongos , Osteoblastos/citologia , Análise de Célula Única
9.
J Geophys Res Atmos ; 121(18): 11104-11118, 2016 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-29082118

RESUMO

Volcanic eruptions are important causes of natural variability in the climate system at all time scales. Assessments of the climate impact of volcanic eruptions by climate models almost universally assume that sulfate aerosol is the only radiatively active volcanic material. We report satellite observations from the Cloud-Aerosol Lidar with Orthogonal Polarization (CALIOP) on board the Cloud-Aerosol Lidar and Infrared Pathfinder Satellite Observations (CALIPSO) satellite after the eruption of Mount Kelud (Indonesia) on 13 February 2014 of volcanic materials in the lower stratosphere. Using these observations along with in situ measurements with the Compact Optical Backscatter AerosoL Detector (COBALD) backscatter sondes and optical particle counters (OPCs) made during a balloon field campaign in northern Australia, we find that fine ash particles with a radius below 0.3 µm likely represented between 20 and 28% of the total volcanic cloud aerosol optical depth 3 months after the eruption. A separation of 1.5-2 km between the ash and sulfate plumes is observed in the CALIOP extinction profiles as well as in the aerosol number concentration measurements of the OPC after 3 months. The settling velocity of fine ash with a radius of 0.3 µm in the tropical lower stratosphere is reduced by 50% due to the upward motion of the Brewer-Dobson circulation resulting a doubling of its lifetime. Three months after the eruption, we find a mean tropical clear-sky radiative forcing at the top of the atmosphere from the Kelud plume near -0.08 W/m2 after including the presence of ash; a value ~20% higher than if sulfate alone is considered. Thus, surface cooling following volcanic eruptions could be affected by the persistence of ash and should be considered in climate simulations.

10.
Stem Cell Reports ; 5(5): 690-701, 2015 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-26455414

RESUMO

Despite advances in our understanding of interactions between mouse hematopoietic stem cells (HSCs) and their niche, little is known about communication between human HSCs and the microenvironment. Using a xenotransplantation model and intravital imaging, we demonstrate that human HSCs display distinct motile behaviors to their hematopoietic progenitor cell (HPC) counterparts, and the same pattern can be found between mouse HSCs and HPCs. HSCs become significantly less motile after transplantation, while progenitor cells remain motile. We show that human HSCs take longer to find their niche than previously expected and suggest that the niche be defined as the position where HSCs stop moving. Intravital imaging is the only technique to determine where in the bone marrow stem cells stop moving, and future analyses should focus on the environment surrounding the HSC at this point.


Assuntos
Movimento Celular , Células-Tronco Hematopoéticas/fisiologia , Osteoblastos/fisiologia , Nicho de Células-Tronco , Animais , Células Cultivadas , Transplante de Células-Tronco Hematopoéticas , Humanos , Camundongos , Camundongos Endogâmicos C57BL
11.
Blood ; 125(8): 1244-55, 2015 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-25573994

RESUMO

Chemokine signaling is important for the seeding of different sites by hematopoietic stem cells (HSCs) during development. Serum response factor (SRF) controls multiple genes governing adhesion and migration, mainly by recruiting members of the myocardin-related transcription factor (MRTF) family of G-actin-regulated cofactors. We used vav-iCre to inactivate MRTF-SRF signaling early during hematopoietic development. In both Srf- and Mrtf-deleted animals, hematopoiesis in fetal liver and spleen is intact but does not become established in fetal bone marrow. Srf-null HSC progenitor cells (HSC/Ps) fail to effectively engraft in transplantation experiments, exhibiting normal proximal signaling responses to SDF-1, but reduced adhesiveness, F-actin assembly, and reduced motility. Srf-null HSC/Ps fail to polarize in response to SDF-1 and cannot migrate through restrictive membrane pores to SDF-1 or Scf in vitro. Mrtf-null HSC/Ps were also defective in chemotactic responses to SDF-1. Srf-null HSC/Ps exhibit substantial deficits in cytoskeletal gene expression. MRTF-SRF signaling is thus critical for expression of genes required for the response to chemokine signaling during hematopoietic development.


Assuntos
Medula Óssea/embriologia , Medula Óssea/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Fator de Resposta Sérica/fisiologia , Nicho de Células-Tronco , Transativadores/fisiologia , Fatores de Transcrição/fisiologia , Animais , Medula Óssea/crescimento & desenvolvimento , Movimento Celular/genética , Células Cultivadas , Regulação da Expressão Gênica no Desenvolvimento , Hematopoese/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais/genética , Nicho de Células-Tronco/genética
12.
Cell Stem Cell ; 13(5): 549-63, 2013 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-24095676

RESUMO

Hematopoietic stem and progenitor cells (HSPCs) are exposed to low levels of oxygen in the bone marrow niche, and hypoxia-inducible factors (HIFs) are the main regulators of cellular responses to oxygen variation. Recent studies using conditional knockout mouse models have unveiled a major role for HIF-1α in the maintenance of murine HSCs; however, the role of HIF-2α is still unclear. Here, we show that knockdown of HIF-2α, and to a much lesser extent HIF-1α, impedes the long-term repopulating ability of human CD34(+) umbilical cord blood cells. HIF-2α-deficient HSPCs display increased production of reactive oxygen species (ROS), which subsequently stimulates endoplasmic reticulum (ER) stress and triggers apoptosis by activation of the unfolded-protein-response (UPR) pathway. HIF-2α deregulation also significantly decreased engraftment ability of human acute myeloid leukemia (AML) cells. Overall, our data demonstrate a key role for HIF-2α in the maintenance of human HSPCs and in the survival of primary AML cells.


Assuntos
Apoptose/fisiologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Leucemia Mieloide Aguda/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Células Cultivadas , Humanos , Leucemia Mieloide Aguda/genética , Camundongos , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo
13.
Cancer Res ; 73(23): 6886-99, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-24145351

RESUMO

Mortality of patients with breast cancer is due overwhelmingly to metastatic spread of the disease. Although dissemination is an early event in breast cancer, extended periods of cancer cell dormancy can result in long latency of metastasis development. Deciphering the mechanisms underlying cancer cell dormancy and subsequent growth at the metastatic site would facilitate development of strategies to interfere with these processes. A challenge in this undertaking has been the lack of models for cancer cell dormancy. We have established novel experimental systems that model the bone microenvironment of the breast cancer metastatic niche. These systems are based on 3D cocultures of breast cancer cells with cell types predominant in bone marrow. We identified conditions in which cancer cells are dormant and conditions in which they proliferate. Dormant cancer cells were able to proliferate upon transfer into supportive microenvironment or upon manipulation of signaling pathways that control dormancy. These experimental systems will be instrumental for metastasis studies, particularly the study of cellular dormancy.


Assuntos
Neoplasias Ósseas/secundário , Neoplasias da Mama/patologia , Modelos Biológicos , Células-Tronco Neoplásicas/fisiologia , Microambiente Tumoral/fisiologia , Animais , Células da Medula Óssea/fisiologia , Pontos de Checagem do Ciclo Celular/fisiologia , Células Cultivadas , Feminino , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Células MCF-7 , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Células-Tronco Neoplásicas/patologia , Nicho de Células-Tronco/fisiologia , Células Estromais/fisiologia
14.
Cell Stem Cell ; 13(2): 161-74, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23910083

RESUMO

In addition to well-characterized CD34(+) hematopoietic stem and progenitor cells (HSPCs), the human hematopoietic stem cell (HSC) hierarchy contains a rare CD34(-) population with severe combined immunodeficiency-repopulating capacity. However, little is known about the molecular characteristics of these CD34(-) cells or their relationship to the CD34(+) populations. Here, we show that the self-renewing Lin(-)CD34(-)CD38(-)CD93(hi) population contains cells that not only function as HSCs, but can also be placed above the CD34(+) populations in the hematopoietic hierarchy. These cells have an active Notch pathway, in which signaling through Delta4 is crucial for maintenance of the primitive state, and combined signals from Jagged1 and TGF-ß are important in controlling its quiescence. They are also refractory to proliferative signals and show a repressed canonical Wnt pathway, in part regulated by Notch. Overall, therefore, CD34(-) cells represent an immature and quiescent human HSC population maintained through a distinctive network of cellular signaling interactions.


Assuntos
Antígenos CD34/metabolismo , Linhagem da Célula , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Transdução de Sinais , ADP-Ribosil Ciclase 1/metabolismo , Células da Medula Óssea/citologia , Diferenciação Celular , Proliferação de Células , Sangue Fetal/citologia , Humanos , Glicoproteínas de Membrana/metabolismo , Receptores de Complemento/metabolismo , Receptores Notch/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Via de Sinalização Wnt
15.
Blood ; 122(10): 1730-40, 2013 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-23814020

RESUMO

Intravital microscopy of the calvarium is the only noninvasive method for high-resolution imaging of the bone marrow (BM) and hematopoietic stem cell (HSC) niches. However, it is unclear if the calvarium is representative of all BM compartments. Using the combination of whole body optical imaging, intravital microscopy, and "in vivo fluorescence trapping," a thorough comparison of HSCs and putative HSC niches in the calvaria, epiphyses, and diaphyses, at steady state or after HSC transplantation, can be made. We report substantial heterogeneity between different BM compartments in terms of bone-remodeling activity (BRA), blood volume fraction (BVF), and hypoxia. Although BVF is high in all BM compartments, including areas adjacent to the endosteum, we found that compartments displaying the highest BVF and BRA were preferentially seeded and engrafted upon HSC transplantation. Unexpectedly, the macroanatomical distribution of HSCs at steady state is homogeneous across these 3 areas and independent of these 2 parameters and suggests the existence of "reconstituting niches," which are distinct from "homeostatic niches." Both types of niches were observed in the calvarium, indicating that endochondral ossification, the process needed for the formation of HSC niches during embryogenesis, is dispensable for the formation of HSC niches during adulthood.


Assuntos
Medula Óssea/anatomia & histologia , Medula Óssea/fisiologia , Compartimento Celular , Células-Tronco Hematopoéticas/citologia , Imageamento Tridimensional/métodos , Animais , Biomarcadores/metabolismo , Vasos Sanguíneos/anatomia & histologia , Vasos Sanguíneos/metabolismo , Volume Sanguíneo , Medula Óssea/irrigação sanguínea , Transplante de Medula Óssea , Remodelação Óssea , Osso e Ossos/irrigação sanguínea , Osso e Ossos/fisiologia , Hipóxia Celular , Células-Tronco Hematopoéticas/metabolismo , Homeostase , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica , Perfusão , Nicho de Células-Tronco
16.
Sci Signal ; 5(235): ra55, 2012 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-22855506

RESUMO

During the early development of the gastrointestinal tract, signaling through the receptor tyrosine kinase RET is required for initiation of lymphoid organ (Peyer's patch) formation and for intestinal innervation by enteric neurons. RET signaling occurs through glial cell line-derived neurotrophic factor (GDNF) family receptor α co-receptors present in the same cell (signaling in cis). It is unclear whether RET signaling in trans, which occurs in vitro through co-receptors from other cells, has a biological role. We showed that the initial aggregation of hematopoietic cells to form lymphoid clusters occurred in a RET-dependent, chemokine-independent manner through adhesion-mediated arrest of lymphoid tissue initiator (LTin) cells. Lymphoid tissue inducer cells were not necessary for this initiation phase. LTin cells responded to all RET ligands in trans, requiring factors from other cells, whereas RET was activated in enteric neurons exclusively by GDNF in cis. Furthermore, genetic and molecular approaches revealed that the versatile RET responses in LTin cells were determined by distinct patterns of expression of the genes encoding RET and its co-receptors. Our study shows that a trans RET response in LTin cells determines the initial phase of enteric lymphoid organ morphogenesis, and suggests that differential co-expression of Ret and Gfra can control the specificity of RET signaling.


Assuntos
Sistema Nervoso Entérico/embriologia , Trato Gastrointestinal/embriologia , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Morfogênese/fisiologia , Nódulos Linfáticos Agregados/embriologia , Proteínas Proto-Oncogênicas c-ret/metabolismo , Transdução de Sinais/fisiologia , Animais , Western Blotting , Células Cultivadas , Trato Gastrointestinal/inervação , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Camundongos , Microscopia Confocal , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Proc Natl Acad Sci U S A ; 107(30): 13414-9, 2010 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-20616004

RESUMO

Thymus organogenesis requires coordinated interactions of multiple cell types, including neural crest (NC) cells, to orchestrate the formation, separation, and subsequent migration of the developing thymus from the third pharyngeal pouch to the thoracic cavity. The molecular mechanisms driving these processes are unclear; however, NC-derived mesenchyme has been shown to play an important role. Here, we show that, in the absence of ephrin-B2 expression on thymic NC-derived mesenchyme, the thymus remains in the cervical area instead of migrating into the thoracic cavity. Analysis of individual NC-derived thymic mesenchymal cells shows that, in the absence of ephrin-B2, their motility is impaired as a result of defective EphB receptor signaling. This implies a NC-derived cell-specific role of EphB-ephrin-B2 interactions in the collective migration of the thymic rudiment during organogenesis.


Assuntos
Efrina-B2/metabolismo , Organogênese , Receptores da Família Eph/metabolismo , Timo/embriologia , Animais , Movimento Celular , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Efrina-B2/genética , Feminino , Citometria de Fluxo , Imuno-Histoquímica , Masculino , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Camundongos Knockout , Microscopia Confocal , Sistema Nervoso/citologia , Sistema Nervoso/embriologia , Sistema Nervoso/metabolismo , Ligação Proteica , Timo/citologia , Timo/inervação
18.
J Immunol ; 184(10): 5686-95, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20404270

RESUMO

Nuclear architecture and chromatin reorganization have recently been shown to orchestrate gene expression and act as key players in developmental pathways. To investigate how regulatory elements in the mouse CD8 gene locus are arranged in space and in relation to each other, three-dimensional fluorescence in situ hybridization and chromosome conformation capture techniques were employed to monitor the repositioning of the locus in relation to its subchromosomal territory and to identify long-range interactions between the different elements during development. Our data demonstrate that CD8 gene expression in murine lymphocytes is accompanied by the relocation of the locus outside its subchromosomal territory. Similar observations in the CD4 locus point to a rather general phenomenon during T cell development. Furthermore, we show that this relocation of the CD8 gene locus is associated with a clustering of regulatory elements forming a tight active chromatin hub in CD8-expressing cells. In contrast, in nonexpressing cells, the gene remains close to the main body of its chromosomal domain and the regulatory elements appear not to interact with each other.


Assuntos
Antígenos CD8/genética , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Núcleo Celular/genética , Regulação da Expressão Gênica no Desenvolvimento/imunologia , Timo/imunologia , Timo/metabolismo , Animais , Antígenos CD4/genética , Antígenos CD8/biossíntese , Posicionamento Cromossômico/genética , Sondas de DNA/genética , Feminino , Imageamento Tridimensional , Hibridização in Situ Fluorescente , Tecido Linfoide/citologia , Tecido Linfoide/imunologia , Tecido Linfoide/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Estrutura Terciária de Proteína/genética , Timo/citologia
19.
Methods Mol Biol ; 616: 161-79, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20379875

RESUMO

This chapter provides information on imaging tools that can be employed to visualise and study lymphoid organ development. We focus on the use of genetically modified mouse models that take advantage of fluorescent protein expression in discrete cell populations, thus allowing live cell imaging during lymphoid organogenesis. We discuss approaches that allow characterisation of the cell types involved in the formation of lymphoid organs, including (i) functional assays in explant organ cultures and (ii) high-resolution whole-mount immunostaining methods, which are useful for the characterisation of specific cell populations in the context of the whole developing organ.


Assuntos
Imageamento Tridimensional/métodos , Sistema Linfático/embriologia , Organogênese , Animais , Linhagem da Célula , Sobrevivência Celular , Dissecação , Embrião de Mamíferos/embriologia , Humanos , Proteínas Luminescentes/metabolismo , Sistema Linfático/citologia , Camundongos , Modelos Animais , Coloração e Rotulagem
20.
J Immunol ; 180(5): 3183-9, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-18292542

RESUMO

Neural crest (NC)-derived mesenchyme has previously been shown to play an important role in the development of fetal thymus. Using Wnt1-Cre and Sox10-Cre mice crossed to Rosa26(eYfp) reporter mice, we have revealed NC-derived mesenchymal cells in the adult murine thymus. We report that NC-derived cells infiltrate the thymus before day 13.5 of embryonic development (E13.5) and differentiate into cells with characteristics of smooth muscle cells associated with large vessels, and pericytes associated with capillaries. In the adult organ at 3 mo of age, these NC-derived perivascular cells continue to be associated with the vasculature, providing structural support to the blood vessels and possibly regulating endothelial cell function.


Assuntos
Senescência Celular/imunologia , Embrião de Mamíferos/citologia , Embrião de Mamíferos/imunologia , Células-Tronco Embrionárias/imunologia , Crista Neural/citologia , Crista Neural/imunologia , Timo/embriologia , Timo/imunologia , Animais , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Senescência Celular/genética , Embrião de Mamíferos/metabolismo , Células-Tronco Embrionárias/metabolismo , Feminino , Desenvolvimento Fetal/genética , Desenvolvimento Fetal/imunologia , Genes Reporter/imunologia , Proteínas Luminescentes/biossíntese , Proteínas Luminescentes/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Crista Neural/metabolismo , Timo/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA