Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Nat Cell Biol ; 25(12): 1736-1745, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38036749

RESUMO

Myeloid cell infiltration of solid tumours generally associates with poor patient prognosis and disease severity1-13. Therefore, understanding the regulation of myeloid cell differentiation during cancer is crucial to counteract their pro-tumourigenic role. Bone marrow (BM) haematopoiesis is a tightly regulated process for the production of all immune cells in accordance to tissue needs14. Myeloid cells differentiate during haematopoiesis from multipotent haematopoietic stem and progenitor cells (HSPCs)15-17. HSPCs can sense inflammatory signals from the periphery during infections18-21 or inflammatory disorders22-27. In these settings, HSPC expansion is associated with increased myeloid differentiation28,29. During carcinogenesis, the elevation of haematopoietic growth factors supports the expansion and differentiation of committed myeloid progenitors5,30. However, it is unclear whether cancer-related inflammation also triggers demand-adapted haematopoiesis at the level of multipotent HSPCs. In the BM, HSPCs reside within the haematopoietic niche which delivers HSC maintenance and differentiation cues31-35. Mesenchymal stem cells (MSCs) are a major cellular component of the BM niche and contribute to HSC homeostasis36-41. Modifications of MSCs in systemic disorders have been associated with HSC differentiation towards myeloid cells22,42. It is unknown if MSCs are regulated in the context of solid tumours and if their myeloid supportive activity is impacted by cancer-induced systemic changes. Here, using unbiased transcriptomic analysis and in situ imaging of HSCs and the BM niche during breast cancer, we show that both HSCs and MSCs are transcriptionally and spatially modified. We demonstrate that breast tumour can distantly remodel the cellular cross-talks in the BM niche leading to increased myelopoiesis.


Assuntos
Medula Óssea , Neoplasias da Mama , Humanos , Feminino , Neoplasias da Mama/patologia , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Multipotentes/metabolismo , Diferenciação Celular , Nicho de Células-Tronco , Células da Medula Óssea
3.
PLoS One ; 16(7): e0254184, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34234374

RESUMO

T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive malignancy with few available targeted therapies. We previously reported that the phosphatase calcineurin (Cn) is required for LIC (leukemia Initiating Capacity) potential of T-ALL pointing to Cn as an interesting therapeutic target. Calcineurin inhibitors have however unwanted side effect. NFAT transcription factors play crucial roles downstream of calcineurin during thymocyte development, T cell differentiation, activation and anergy. Here we elucidate NFAT functional relevance in T-ALL. Using murine T-ALL models in which Nfat genes can be inactivated either singly or in combination, we show that NFATs are required for T-ALL LIC potential and essential to survival, proliferation and migration of T-ALL cells. We also demonstrate that Nfat genes are functionally redundant in T-ALL and identified a node of genes commonly deregulated upon Cn or NFAT inactivation, which may serve as future candidate targets for T-ALL.


Assuntos
Fatores de Transcrição NFATC/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Linfócitos T/metabolismo , Animais , Calcineurina/metabolismo , Inibidores de Calcineurina/farmacologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Linfócitos T/efeitos dos fármacos
4.
Front Cell Dev Biol ; 9: 645496, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33996805

RESUMO

The bone marrow (BM) tissue is the main physiological site for adult hematopoiesis. In recent years, the cellular and matrix components composing the BM have been defined with unprecedent resolution, both at the molecular and structural levels. With the expansion of this knowledge, the possibility of reproducing a BM-like structure, to ectopically support and study hematopoiesis, becomes a reality. A number of experimental systems have been implemented and have displayed the feasibility of bioengineering BM tissues, supported by cells of mesenchymal origin. Despite being known as an abundant component of the BM, the vasculature has been largely disregarded for its role in regulating tissue formation, organization and determination. Recent reports have highlighted the crucial role for vascular endothelial cells in shaping tissue development and supporting steady state, emergency and malignant hematopoiesis, both pre- and postnatally. Herein, we review the field of BM-tissue bioengineering with a particular focus on vascular system implementation and integration, starting from describing a variety of applicable in vitro models, ending up with in vivo preclinical models. Additionally, we highlight the challenges of the field and discuss the clinical perspectives in terms of adoptive transfer of vascularized BM-niche grafts in patients to support recovering hematopoiesis.

5.
Cell Rep ; 35(6): 109119, 2021 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-33979628

RESUMO

The bone-marrow (BM) niche is the spatial environment composed by a network of multiple stromal components regulating adult hematopoiesis. We use multi-omics and computational tools to analyze multiple BM environmental compartments and decipher their mutual interactions in the context of acute myeloid leukemia (AML) xenografts. Under homeostatic conditions, we find a considerable overlap between niche populations identified using current markers. Our analysis defines eight functional clusters of genes informing on the cellular identity and function of the different subpopulations and pointing at specific stromal interrelationships. We describe how these transcriptomic profiles change during human AML development and, by using a proximity-based molecular approach, we identify early disease onset deregulated genes in the mesenchymal compartment. Finally, we analyze the BM proteomic secretome in the presence of AML and integrate it with the transcriptome to predict signaling nodes involved in niche alteration in AML.


Assuntos
Células da Medula Óssea/metabolismo , Leucemia Mieloide Aguda/genética , Proteômica/métodos , Animais , Humanos , Camundongos , Microambiente Tumoral
7.
Nat Protoc ; 16(2): 872-892, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33311715

RESUMO

Understanding cell-cell interactions is critical in most, if not all, research fields in biology. Nevertheless, studying intercellular crosstalk in vivo remains a relevant challenge, due mainly to the difficulty in spatially locating the surroundings of particular cells in the tissue. Cherry-niche is a powerful new method that enables cells expressing a fluorescent protein to label their surrounding cells, facilitating their specific isolation from the whole tissue as live cells. We previously applied Cherry-niche in cancer research to study the tumor microenvironment (TME) in metastasis. Here we describe how to generate cancer cells with the ability to label their neighboring cells (within the tumor niche) by transferring a liposoluble fluorescent protein. Live niche cells can be isolated and compared with cells distant from the tumor bulk, using a variety of ex vivo approaches. As previously shown, this system has the potential to identify novel components in the TME and improve our understanding of their local interactions. Importantly, Cherry-niche can also be applied to study potential cell-cell interactions due to in vivo proximity in research fields beyond cancer. This protocol takes 2-3 weeks to generate the labeling cells and 1-2 weeks to test their labeling ability.


Assuntos
Comunicação Celular/fisiologia , Imuno-Histoquímica/métodos , Corantes Fluorescentes/química , Humanos , Neoplasias/imunologia , Neoplasias/patologia , Microambiente Tumoral/imunologia , Microambiente Tumoral/fisiologia
8.
Cancer Cell ; 38(6): 753-756, 2020 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-33321083

RESUMO

Imaging has had a profound impact on our ability to understand and treat cancer. We invited some experts to discuss imaging approaches that can be used in various aspects of cancer research, from investigating the complexity and diversity of cancer cells and their environments to guiding clinical decision-making.


Assuntos
Imageamento Tridimensional/métodos , Imagem Molecular/métodos , Neoplasias/diagnóstico , Humanos , Terapia de Alvo Molecular , Neoplasias/genética , Neoplasias/metabolismo , Medicina de Precisão , Análise de Célula Única , Microambiente Tumoral
9.
Cell Stem Cell ; 27(5): 702-704, 2020 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-33157046

RESUMO

COVID-19 has unfortunately halted lab work, conferences, and in-person networking, which is especially detrimental to researchers just starting their labs. Through social media and our reviewer networks, we met some early-career stem cell investigators impacted by the closures. Here, they introduce themselves and their research to our readers.

11.
Nat Cell Biol ; 22(2): 257, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31969686

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

12.
Nat Cell Biol ; 22(1): 7-17, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31907409

RESUMO

Adult haematopoietic stem cells (HSCs) mainly reside in the bone marrow, where stromal and haematopoietic cells regulate their function. The steady state HSC niche has been extensively studied. In this Review, we focus on how bone marrow microenvironment components respond to different insults including inflammation, malignant haematopoiesis and chemotherapy. We highlight common and unique patterns among multiple cell types and their environment and discuss current limitations in our understanding of this complex and dynamic tissue.


Assuntos
Diferenciação Celular/fisiologia , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Nicho de Células-Tronco/fisiologia , Animais , Medula Óssea/metabolismo , Meio Ambiente , Humanos
14.
J Exp Med ; 215(3): 729-743, 2018 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-29453226

RESUMO

Xenotransplantation of patient-derived samples in mouse models has been instrumental in depicting the role of hematopoietic stem and progenitor cells in the establishment as well as progression of hematological malignancies. The foundations for this field of research have been based on the development of immunodeficient mouse models, which provide normal and malignant human hematopoietic cells with a supportive microenvironment. Immunosuppressed and genetically modified mice expressing human growth factors were key milestones in patient-derived xenograft (PDX) models, highlighting the importance of developing humanized microenvironments. The latest major improvement has been the use of human bone marrow (BM) niche-forming cells to generate human-mouse chimeric BM tissues in PDXs, which can shed light on the interactions between human stroma and hematopoietic cells. Here, we summarize the methods used for human hematopoietic cell xenotransplantation and their milestones and review the latest approaches in generating humanized BM tissues in mice to study human normal and malignant hematopoiesis.


Assuntos
Bioengenharia/métodos , Transplante de Medula Óssea , Medula Óssea/metabolismo , Nicho de Células-Tronco , Animais , Hematopoese , Humanos , Camundongos , Transplante Heterólogo
15.
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
16.
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
17.
Stem Cell Reports ; 9(1): 5-11, 2017 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-28648895

RESUMO

Nuclear factor erythroid-derived 2 (NF-E2) has been associated with megakaryocyte maturation and platelet production. Recently, an increased in NF-E2 activity has been implicated in myeloproliferative neoplasms. Here, we investigate the role of NF-E2 in normal human hematopoiesis. Knockdown of NF-E2 in the hematopoietic stem and progenitor cells (HSPCs) not only reduced the formation of megakaryocytes but also drastically impaired hematopoietic stem cell activity, decreasing human engraftment in immunodeficient (NSG) mice. This phenotype is likely to be related to both increased cell proliferation (p21-mediated) and reduced Notch1 protein expression, which favors HSPC differentiation over self-renewal. Strikingly, although NF-E2 silencing in HSPCs did not affect their myeloid and B cell differentiation in vivo, it almost abrogated T cell production in primary hosts, as confirmed by in vitro studies. This effect is at least partly due to Notch1 downregulation in NF-E2-silenced HSPCs. Together these data reveal that NF-E2 is an important driver of human hematopoietic stem cell maintenance and T lineage differentiation.


Assuntos
Proliferação de Células , Células-Tronco Hematopoéticas/citologia , Linfopoese , Fator de Transcrição NF-E2/metabolismo , Receptor Notch1/metabolismo , Linfócitos T/citologia , Animais , Linhagem Celular , Células Cultivadas , Regulação da Expressão Gênica , Inativação Gênica , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Humanos , Camundongos , Camundongos SCID , Fator de Transcrição NF-E2/genética , Receptor Notch1/genética , Linfócitos T/metabolismo
18.
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
19.
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
20.
Immunol Rev ; 271(1): 156-72, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27088913

RESUMO

Intensive chemotherapy regimens have led to a substantial improvement in the cure rate of patients suffering from T-cell acute lymphoblastic leukemia (T-ALL). Despite this progress, about 15% and 50% of pediatric and adult cases, respectively, show resistance to treatment or relapse with dismal prognosis, calling for further therapeutic investigations. T-ALL is an heterogeneous disease, which presents intrinsic alterations leading to aberrant expression of transcription factors normally involved in hematopoietic stem/progenitor cell development and mutations in genes implicated in the regulation of cell cycle progression, apoptosis, and T-cell development. Gene expression profiling allowed the classification of T-ALL into defined molecular subgroups that mostly reflects the stage of their differentiation arrest. So far this knowledge has not translated into novel, targeted therapy. Recent evidence points to the importance of extrinsic signaling cues in controlling the ability of T-ALL to home, survive, and proliferate, thus offering the perspective of new therapeutic options. This review summarizes the present understanding of the interactions between hematopoietic cells and bone marrow/thymic niches during normal hematopoiesis, describes the main signaling pathways implicated in this dialog, and finally highlights how malignant T cells rely on specific niches to maintain their ability to sustain and propagate leukemia.


Assuntos
Medula Óssea/imunologia , Carcinogênese , Leucemia-Linfoma Linfoblástico de Células T Precursoras/imunologia , Linfócitos T/fisiologia , Timo/imunologia , Adulto , Animais , Diferenciação Celular , Microambiente Celular , Criança , Hematopoese , Humanos , Terapia de Alvo Molecular , Transdução de Sinais , Transcriptoma
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA