Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Haematologica ; 106(1): 74-86, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31949009

RESUMO

Allogeneic hematopoietic stem cell transplantation is the treatment of choice for autosomal recessive osteopetrosis caused by defects in the TCIRG1 gene. Despite recent progress in conditioning, a relevant number of patients are not eligible for allogeneic stem cell transplantation because of the severity of the disease and significant transplant-related morbidity. We exploited peripheral CD34+ cells, known to circulate at high frequency in the peripheral blood of TCIRG1-deficient patients, as a novel cell source for autologous transplantation of gene corrected cells. Detailed phenotypical analysis showed that circulating CD34+ cells have a cellular composition that resembles bone marrow, supporting their use in gene therapy protocols. Transcriptomic profile revealed enrichment in genes expressed by hematopoietic stem and progenitor cells (HSPCs). To overcome the limit of bone marrow harvest/ HSPC mobilization and serial blood drawings in TCIRG1 patients, we applied UM171-based ex-vivo expansion of HSPCs coupled with lentiviral gene transfer. Circulating CD34+ cells from TCIRG1-defective patients were transduced with a clinically-optimized lentiviral vector (LV) expressing TCIRG1 under the control of phosphoglycerate promoter and expanded ex vivo. Expanded cells maintained long-term engraftment capacity and multi-lineage repopulating potential when transplanted in vivo both in primary and secondary NSG recipients. Moreover, when CD34+ cells were differentiated in vitro, genetically corrected osteoclasts resorbed the bone efficiently. Overall, we provide evidence that expansion of circulating HSPCs coupled to gene therapy can overcome the limit of stem cell harvest in osteopetrotic patients, thus opening the way to future gene-based treatment of skeletal diseases caused by bone marrow fibrosis.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Osteopetrose , ATPases Vacuolares Próton-Translocadoras , Antígenos CD34 , Terapia Genética , Células-Tronco Hematopoéticas/metabolismo , Humanos , Osteoclastos/metabolismo , Osteopetrose/genética , Osteopetrose/terapia , ATPases Vacuolares Próton-Translocadoras/genética , ATPases Vacuolares Próton-Translocadoras/metabolismo
2.
Stem Cells ; 37(7): 876-887, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30895693

RESUMO

In spite of the progress in gene editing achieved in recent years, a subset of genetic diseases involving structural chromosome abnormalities, including aneuploidies, large deletions and complex rearrangements, cannot be treated with conventional gene therapy approaches. We have previously devised a strategy, dubbed chromosome transplantation (CT), to replace an endogenous mutated chromosome with an exogenous normal one. To establish a proof of principle for our approach, we chose as disease model the chronic granulomatous disease (CGD), an X-linked severe immunodeficiency due to abnormalities in CYBB (GP91) gene, including large genomic deletions. We corrected the gene defect by CT in induced pluripotent stem cells (iPSCs) from a CGD male mouse model. The Hprt gene of the endogenous X chromosome was inactivated by CRISPR/Cas9 technology thus allowing the exploitation of the hypoxanthine-aminopterin-thymidine selection system to introduce a normal donor X chromosome by microcell-mediated chromosome transfer. X-transplanted clones were obtained, and diploid XY clones which spontaneously lost the endogenous X chromosome were isolated. These cells were differentiated toward the myeloid lineage, and functional granulocytes producing GP91 protein were obtained. We propose the CT approach to correct iPSCs from patients affected by other X-linked diseases with large deletions, whose treatment is still unsatisfactory. Stem Cells 2019;37:876-887.


Assuntos
Cromossomos de Mamíferos , Terapia Genética/métodos , Granulócitos/metabolismo , Doença Granulomatosa Crônica/terapia , Hipoxantina Fosforribosiltransferase/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , NADPH Oxidase 2/genética , Aminopterina/metabolismo , Aminopterina/farmacologia , Animais , Sequência de Bases , Sistemas CRISPR-Cas , Diferenciação Celular , Células Clonais , Meios de Cultura/química , Modelos Animais de Doenças , Edição de Genes/métodos , Granulócitos/citologia , Granulócitos/efeitos dos fármacos , Doença Granulomatosa Crônica/genética , Doença Granulomatosa Crônica/metabolismo , Doença Granulomatosa Crônica/patologia , Humanos , Hipoxantina/metabolismo , Hipoxantina/farmacologia , Hipoxantina Fosforribosiltransferase/deficiência , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/patologia , Masculino , Camundongos , NADPH Oxidase 2/deficiência , Estudo de Prova de Conceito , Deleção de Sequência , Tioguanina/metabolismo , Tioguanina/farmacologia , Timidina/metabolismo , Timidina/farmacologia , Cromossomo X/química , Cromossomo X/metabolismo
3.
Haematologica ; 104(9): 1744-1755, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30792210

RESUMO

The balance between self-renewal and differentiation is crucial to ensure the homeostasis of the hematopoietic system, and is a hallmark of hematopoietic stem cells. However, the underlying molecular pathways, including the role of micro-RNA, are not completely understood. To assess the contribution of micro-RNA, we performed micro-RNA profiling of hematopoietic stem cells and their immediate downstream progeny multi-potent progenitors from wild-type control and Pbx1-conditional knockout mice, whose stem cells display a profound self-renewal defect. Unsupervised hierarchical cluster analysis separated stem cells from multi-potent progenitors, suggesting that micro-RNA might regulate the first transition step in the adult hematopoietic development. Notably, Pbx1-deficient and wild-type cells clustered separately, linking micro-RNAs to self-renewal impairment. Differential expression analysis of micro-RNA in the physiological stem cell-to-multi-potent progenitor transition and in Pbx1-deficient stem cells compared to control stem cells revealed miR-127-3p as the most differentially expressed. Furthermore, miR-127-3p was strongly stem cell-specific, being quickly down-regulated upon differentiation and not re-expressed further downstream in the bone marrow hematopoietic hierarchy. Inhibition of miR-127-3p function in Lineage-negative cells, achieved through a lentiviral-sponge vector, led to severe stem cell depletion, as assessed with serial transplantation assays. miR-127-3p-sponged stem cells displayed accelerated differentiation, which was uncoupled from proliferation, accounting for the observed stem cell reduction. miR-127-3p overexpression in Lineage-negative cells did not alter stem cell pool size, but gave rise to lymphopenia, likely due to lack of miR-127-3p physiological downregulation beyond the stem cell stage. Thus, tight regulation of miR-127-3p is crucial to preserve the self-renewing stem cell pool and homeostasis of the hematopoietic system.


Assuntos
Diferenciação Celular , Células-Tronco Hematopoéticas/citologia , MicroRNAs/fisiologia , Animais , Linhagem da Célula/genética , Análise por Conglomerados , Cruzamentos Genéticos , Perfilação da Expressão Gênica , Hematopoese , Homeostase , Humanos , Células K562 , Lentivirus/genética , Camundongos , Camundongos Knockout , Estresse Oxidativo , Fator de Transcrição 1 de Leucemia de Células Pré-B/metabolismo
4.
J Cell Sci ; 126(Pt 14): 3181-91, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23660001

RESUMO

The capacity of the hematopoietic system to promptly respond to peripheral demands relies on adequate pools of progenitors able to transiently proliferate and differentiate in a regulated manner. However, little is known about factors that may restrain progenitor maturation to maintain their reservoirs. Conditional knockout mice for the Pbx1 proto-oncogene have a significant reduction in lineage-restricted progenitors in addition to a profound defect in hematopoietic stem cell (HSC) self-renewal. Through analysis of purified progenitor proliferation, differentiation capacity and transcriptional profiling, we demonstrate that Pbx1 regulates the lineage-specific output of multipotent and oligopotent progenitors. In the absence of Pbx1 multipotent progenitor (MPP) and common myeloid progenitor (CMP) pools are reduced due to aberrantly rapid myeloid maturation. This is associated with premature expression of myeloid differentiation genes and decreased maintenance of proto-oncogene transcriptional pathways, including reduced expression of Meis1, a Pbx1 dimerization partner, and its subordinate transcriptional program. Conversely, Pbx1 maintains the lymphoid differentiation potential of lymphoid-primed MPPs (LMPPs) and common lymphoid progenitors (CLPs), whose reduction in the absence of Pbx1 is associated with a defect in lymphoid priming that is also present in CMPs, which persistently express lymphoid and HSC genes underlying a previously unappreciated lineage promiscuity that is maintained by Pbx1. These results demonstrate a role for Pbx1 in restraining myeloid maturation while maintaining lymphoid potential to appropriately regulate progenitor reservoirs.


Assuntos
Hematopoese , Proteínas de Homeodomínio/metabolismo , Células Progenitoras Linfoides/fisiologia , Células Progenitoras Mieloides/fisiologia , Proteínas de Neoplasias/metabolismo , Fatores de Transcrição/metabolismo , Animais , Linhagem da Célula/genética , Sobrevivência Celular/genética , Células Cultivadas , Regulação da Expressão Gênica , Proteínas de Homeodomínio/genética , Ativação Linfocitária/genética , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Proteína Meis1 , Fator de Transcrição 1 de Leucemia de Células Pré-B , Multimerização Proteica , Fatores de Transcrição/genética , Ativação Transcricional
5.
Blood ; 120(5): 1005-14, 2012 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-22723555

RESUMO

Omenn syndrome (OS) is an atypical primary immunodeficiency characterized by severe autoimmunity because of activated T cells infiltrating target organs. The impaired recombinase activity in OS severely affects expression of the pre-T-cell receptor complex in immature thymocytes, which is crucial for an efficient development of the thymic epithelial component. Anti-CD3ε monoclonal antibody (mAb) treatment in RAG2(-/-) mice was previously shown to mimic pre-TCR signaling promoting thymic expansion. Here we show the effect of anti-CD3ε mAb administration in the RAG2(R229Q) mouse model, which closely recapitulates human OS. These animals, in spite of the inability to induce the autoimmune regulator, displayed a significant amelioration in thymic epithelial compartment and an important reduction of peripheral T-cell activation and tissue infiltration. Furthermore, by injecting a high number of RAG2(R229Q) progenitors into RAG2(-/-) animals previously conditioned with anti-CD3ε mAb, we detected autoimmune regulator expression together with the absence of peripheral immunopathology. These observations indicate that improving epithelial thymic function might ameliorate the detrimental behavior of the cell-autonomous RAG defect. Our data provide important therapeutic proof of concept for future clinical applications of anti-CD3ε mAb treatment in severe combined immunodeficiency forms characterized by poor thymus function and autoimmunity.


Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Doenças Autoimunes/prevenção & controle , Complexo CD3/imunologia , Imunodeficiência Combinada Severa/terapia , Timo/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Autoimunidade/efeitos dos fármacos , Autoimunidade/genética , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tamanho do Órgão/efeitos dos fármacos , Imunodeficiência Combinada Severa/imunologia , Imunodeficiência Combinada Severa/patologia , Timo/imunologia , Timo/patologia , Timo/ultraestrutura
6.
Front Cell Dev Biol ; 12: 1372873, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38404687

RESUMO

Pre-B cell leukemia factor 1 (PBX1) is a Three Aminoacid Loop Extension (TALE) homeodomain-containing transcription factor playing crucial roles in organ pattering during embryogenesis, through the formation of nuclear complexes with other TALE class and/or homeobox proteins to regulate target genes. Its contribution to the development of several organs has been elucidated mainly through the study of murine knockout models. A crucial role for human development has been recently highlighted through the discovery of different de novo pathogenic PBX1 variants in children affected by developmental defects. In the adult, PBX1 is expressed in selected tissues such as in the brain, in the gastro-intestinal and urinary systems, or in hematopoietic stem and progenitor cells, while in other organs is barely detectable. When involved in the t(1;19) chromosomal translocation it acts as an oncogene, since the resulting fusion protein drives pre-B cell leukemia, due to the induction of target genes not normally targeted by the native protein. Its aberrant expression has been associated to tumor development, progression, or therapy-resistance as in breast cancer, ovarian cancer or myeloproliferative neoplasm (MPN). On the other hand, in colorectal cancer PBX1 functions as a tumor suppressor, highlighting its context-dependent role. We here discuss differences and analogies of PBX1 roles during embryonic development and in cancer, focusing mainly on the most recent discoveries.

7.
J Cell Sci ; 124(Pt 13): 2208-19, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21670200

RESUMO

The mixed lineage leukemia (MLL) proto-oncogenic protein is a histone-lysine N-methyltransferase that is produced by proteolytic cleavage and self-association of the respective functionally distinct subunits (MLL(N) and MLL(C)) to form a holocomplex involved in epigenetic transcriptional regulation. On the basis of studies in Drosophila it has been suggested that the separated subunits might also have distinct functions. In this study, we used a genetically engineered mouse line that lacked MLL(C) to show that the MLL(N)-MLL(C) holocomplex is responsible for MLL functions in various developmental processes. The stability of MLL(N) is dependent on its intramolecular interaction with MLL(C), which is mediated through the first and fourth plant homeodomain (PHD) fingers (PHD1 and PHD4) and the phenylalanine/tyrosine-rich (FYRN) domain of MLL(N). Free MLL(N) is destroyed by a mechanism that targets the FYRN domain, whereas free MLL(C) is exported to the cytoplasm and degraded by the proteasome. PHD1 is encoded by an alternatively spliced exon that is occasionally deleted in T-cell leukemia, and its absence produces an MLL mutant protein that is deficient for holocomplex formation. Therefore, this should be a loss-of-function mutant allele, suggesting that the known tumor suppression role of MLL may also apply to the T-cell lineage. Our data demonstrate that the dissociated MLL subunits are subjected to distinct degradation pathways and thus not likely to have separate functions unless the degradation mechanisms are inhibited.


Assuntos
Proteína de Leucina Linfoide-Mieloide/metabolismo , Animais , Linhagem Celular , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Leucemia de Células T/genética , Camundongos , Camundongos Knockout , Mutação , Proteína de Leucina Linfoide-Mieloide/genética , Proteínas do Grupo Polycomb , Processamento de Proteína Pós-Traducional , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica
8.
J Emerg Manag ; 21(6): 557-576, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38189205

RESUMO

Understanding disasters as socially constructed events represents a departure from current and historic ways in which disasters are characterized, requiring a focal shift in thinking from forces of nature toward social order. Changing societal reactions to evolving natural occurrences restores disasters within the social order, introducing law as an essential framework in approaching disasters as injustices as opposed to misfortunes. International attention is starting to shift strategies intended to reduce risks to natural or man-made hazards and increasing attention on methods toward minimizing their impact known as disaster risk reduction (DRR). DRR is "a policy aimed at preventing new and reducing existing disaster risk and managing residual risk, all of which contribute to strengthening resilience and therefore to the achievement of sustainable development." The development of normative frameworks to reinforce disaster governance is a significant component in enhancing disaster management systems. Disaster law is an emerging tool to regulate "governance, ethics, and decisions on the demands of a sustainable, inclusive, and healthy planet." International legal frameworks heavily influence disaster prevention and preparedness with an increased central focus on implementing International Human Rights Law in DRR practices. Legal structures protecting human rights in DRR initiatives positively obligate states to take proper and necessary actions to prevent harm from future disasters. The application of human rights standards fosters the paradigm shift from evaluation of the hazards impact toward assessments of states' negligence of risks. Interactions among the natural environment, socio-demographics, and the built environment are strong predictors for disaster losses, thus "the regulatory potential for avoiding disasters and reducing their consequence is obvious." Preventative action becomes a crucial element if the catalyst of the disaster event is failure to adequately prepare and social vulnerability. Disaster law encompasses participation, damage control, and local habitat management as mandatory conditions of governance, assigning criminal liability to public administrators' negligence toward disaster planning and/or enforcement. Disaster law produces a "sustainable, reliable, and cost-effective model for addressing disasters," empowering communities to participate in disaster management efforts, one of the strongest methods of building resilience and reducing risk to disasters.


Assuntos
Planejamento em Desastres , Desastres , Humanos , Pessoal Administrativo , Direitos Humanos
9.
Biomedicines ; 11(10)2023 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-37892987

RESUMO

Inflammation impacts human hematopoiesis across physiologic and pathologic conditions, as signals derived from the bone marrow microenvironment, such as pro-inflammatory cytokines and chemokines, have been shown to alter hematopoietic stem cell (HSCs) homeostasis. Dysregulated inflammation can skew HSC fate-related decisions, leading to aberrant hematopoiesis and potentially contributing to the pathogenesis of hematological disorders such as myelodysplastic syndromes (MDS). Recently, emerging studies have used single-cell sequencing and muti-omic approaches to investigate HSC cellular heterogeneity and gene expression in normal hematopoiesis as well as in myeloid malignancies. This review summarizes recent reports mechanistically dissecting the role of inflammatory signaling and innate immune response activation due to MDS progression. Furthermore, we highlight the growing importance of using multi-omic techniques, such as single-cell profiling and deconvolution methods, to unravel MDSs' heterogeneity. These approaches have provided valuable insights into the patterns of clonal evolution that drive MDS progression and have elucidated the impact of inflammation on the composition of the bone marrow immune microenvironment in MDS.

10.
Wiley Interdiscip Rev RNA ; 13(3): e1693, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34532984

RESUMO

The hematopoietic system is sustained over time by a small pool of hematopoietic stem cells (HSCs). They reside at the apex of a complex hierarchy composed of cells with progressively more restricted lineage potential, regenerative capacity, and with different proliferation characteristics. Like other somatic stem cells, HSCs are endowed with long-term self-renewal and multipotent differentiation ability, to sustain the high turnover of mature cells such as erythrocytes or granulocytes, and to rapidly respond to acute peripheral stresses including bleeding, infections, or inflammation. Maintenance of both attributes over time, and of the proper balance between these opposite features, is crucial to ensure the homeostasis of the hematopoietic system. Micro-RNAs (miRNAs) are short non-coding RNAs that regulate gene expression posttranscriptionally upon binding to specific mRNA targets. In the past 10 years they have emerged as important players for preserving the HSC pool by acting on several biological mechanisms, such as maintenance of the quiescent state while preserving proliferation ability, prevention of apoptosis, premature differentiation, lineage skewing, excessive expansion, or retention within the BM niche. miRNA-mediated posttranscriptional fine-tuning of all these processes constitutes a safety mechanism to protect HSCs, by complementing the action of transcription factors and of other regulators and avoiding unwanted expansion or aplasia. The current knowledge of miRNAs function in different aspects of HSC biology, including consequences of aberrant miRNA expression, will be reviewed; yet unsolved issues will be discussed. This article is categorized under: RNA in Disease and Development > RNA in Disease RNA in Disease and Development > RNA in Development.


Assuntos
MicroRNAs , Apoptose , Diferenciação Celular/genética , Células-Tronco Hematopoéticas/metabolismo , Homeostase , MicroRNAs/genética , MicroRNAs/metabolismo
11.
Cells ; 11(3)2022 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-35159366

RESUMO

Inherited blood disorders comprise a large spectrum of diseases due to germline mutations in genes with key function in the hematopoietic system; they include immunodeficiencies, anemia or metabolic diseases. For most of them the only curative treatment is bone marrow transplantation, a procedure associated to severe complications; other therapies include red blood cell and platelet transfusions, which are dependent on donor availability. An alternative option is gene therapy, in which the wild-type form of the mutated gene is delivered into autologous hematopoietic stem cells using viral vectors. A more recent therapeutic perspective is gene correction through CRISPR/Cas9-mediated gene editing, that overcomes safety concerns due to insertional mutagenesis and allows correction of base substitutions in large size genes difficult to incorporate into vectors. However, applying this technique to genomic disorders caused by large gene deletions is challenging. Chromosomal transplantation has been proposed as a solution, using a universal source of wild-type chromosomes as donor, and induced pluripotent stem cells (iPSCs) as acceptor. One of the obstacles to be addressed for translating PSC research into clinical practice is the still unsatisfactory differentiation into transplantable hematopoietic stem or mature cells. We provide an overview of the recent progresses in this field and discuss challenges and potential of iPSC-based therapies for the treatment of inherited blood disorders.


Assuntos
Transtornos Herdados da Coagulação Sanguínea/sangue , Edição de Genes/métodos , Terapia Genética/métodos , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Pluripotentes/metabolismo , Diferenciação Celular , Humanos
12.
Front Immunol ; 13: 884024, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35603212

RESUMO

The bone marrow (BM) niche is the spatial structure within the intra-trabecular spaces of spongious bones and of the cavity of long bones where adult haematopoietic stem cells (HSCs) maintain their undifferentiated and cellular self-renewal state through the intervention of vascular and nervous networks, metabolic pathways, transcriptional and epigenetic regulators, and humoral signals. Within the niche, HSCs interact with various cell types such as osteoblasts, endothelial cells, macrophages, and mesenchymal stromal cells (MSCs), which maintain HSCs in a quiescent state or sustain their proliferation, differentiation, and trafficking, depending on body needs. In physiological conditions, the BM niche permits the daily production of all the blood and immune cells and their admittance/ingress/progression into the bloodstream. However, disruption of this delicate microenvironment promotes the initiation and progression of malignancies such as those included in the spectrum of myeloid neoplasms, also favouring resistance to pharmacological therapies. Alterations in the MSC population and in the crosstalk with HSCs owing to tumour-derived factors contribute to the formation of a malignant niche. On the other hand, cells of the BM microenvironment cooperate in creating a unique milieu favouring metastasization of distant tumours into the bone. In this framework, the pro-tumorigenic role of MSCs is well-documented, and few evidence suggest also an anti-tumorigenic effect. Here we will review recent advances regarding the BM niche composition and functionality in normal and in malignant conditions, as well as the therapeutic implications of the interplay between its diverse cellular components and malignant cells.


Assuntos
Medula Óssea , Células-Tronco Mesenquimais , Medula Óssea/metabolismo , Células Endoteliais , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Nicho de Células-Tronco
13.
Stem Cell Reports ; 16(11): 2607-2616, 2021 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-34678207

RESUMO

PBX1 regulates the balance between self-renewal and differentiation of hematopoietic stem cells and maintains proto-oncogenic transcriptional pathways in early progenitors. Its increased expression was found in myeloproliferative neoplasm (MPN) patients bearing the JAK2V617F mutation. To investigate if PBX1 contributes to MPN, and to explore its potential as therapeutic target, we generated the JP mouse strain, in which the human JAK2 mutation is induced in the absence of PBX1. Typical MPN features, such as thrombocythemia and granulocytosis, did not develop without PBX1, while erythrocytosis, initially displayed by JP mice, gradually resolved over time; splenic myeloid metaplasia and in vitro cytokine independent growth were absent upon PBX1 inactivation. The aberrant transcriptome in stem/progenitor cells from the MPN model was reverted by the absence of PBX1, demonstrating that PBX1 controls part of the molecular pathways deregulated by the JAK2V617F mutation. Modulation of the PBX1-driven transcriptional program might represent a novel therapeutic approach.


Assuntos
Regulação Neoplásica da Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Transtornos Mieloproliferativos/genética , Neoplasias/genética , Fator de Transcrição 1 de Leucemia de Células Pré-B/genética , Animais , Modelos Animais de Doenças , Progressão da Doença , Perfilação da Expressão Gênica/métodos , Humanos , Camundongos Knockout , Camundongos Transgênicos , Mutação , Transtornos Mieloproliferativos/metabolismo , Transtornos Mieloproliferativos/patologia , Neoplasias/metabolismo , Neoplasias/patologia , Fator de Transcrição 1 de Leucemia de Células Pré-B/metabolismo , RNA-Seq/métodos , Transdução de Sinais/genética
14.
Bone Rep ; 12: 100242, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31938717

RESUMO

BACKGROUND: Autosomal recessive osteopetrosis is a rare skeletal disorder with increased bone density due to a failure in osteoclast bone resorption. In most cases, the defect is cell-autonomous, and >50% of patients bear mutations in the TCIRG1 gene, encoding for a subunit of the vacuolar proton pump essential for osteoclast resorptive activity. The only cure is hematopoietic stem cell transplantation, which corrects the bone pathology by allowing the formation of donor-derived functional osteoclasts. Therapeutic approaches using patient-derived cells corrected ex vivo through viral transduction or gene editing can be considered, but to date functional rescue cannot be demonstrated in vivo because a relevant animal model for xenotransplant is missing. METHODS: We generated a new mouse model, which we named NSG oc/oc, presenting severe autosomal recessive osteopetrosis owing to the Tcirg1 oc mutation, and profound immunodeficiency caused by the NSG background. We performed neonatal murine bone marrow transplantation and xenotransplantation with human CD34+ cells. RESULTS: We demonstrated that neonatal murine bone marrow transplantation rescued NSG oc/oc mice, in line with previous findings in the oc/oc parental strain and with evidence from clinical practice in humans. Importantly, we also demonstrated human cell chimerism in the bone marrow of NSG oc/oc mice transplanted with human CD34+ cells. The severity and rapid progression of the disease in the mouse model prevented amelioration of the bone pathology; nevertheless, we cannot completely exclude that minor early modifications of the bone tissue might have occurred. CONCLUSION: Our work paves the way to generating an improved xenograft model for in vivo evaluation of functional rescue of patient-derived corrected cells. Further refinement of the newly generated mouse model will allow capitalizing on it for an optimized exploitation in the path to novel cell therapies.

15.
Nat Commun ; 9(1): 676, 2018 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-29445158

RESUMO

Atypical chemokine receptors (ACKRs) are regulators of leukocyte traffic, inflammation, and immunity. ACKR2 is a scavenger for most inflammatory CC chemokines and is a negative regulator of inflammation. Here we report that ACKR2 is expressed in hematopoietic precursors and downregulated during myeloid differentiation. Genetic inactivation of ACKR2 results in increased levels of inflammatory chemokine receptors and release from the bone marrow of neutrophils with increased anti-metastatic activity. In a model of NeuT-driven primary mammary carcinogenesis ACKR2 deficiency is associated with increased primary tumor growth and protection against metastasis. ACKR2 deficiency results in neutrophil-mediated protection against metastasis in mice orthotopically transplanted with 4T1 mammary carcinoma and intravenously injected with B16F10 melanoma cell lines. Thus, ACKR2 is a key regulator (checkpoint) of mouse myeloid differentiation and function and its targeting unleashes the anti-metastatic activity of neutrophils in mice.


Assuntos
Células-Tronco Hematopoéticas/metabolismo , Neoplasias Experimentais/metabolismo , Neutrófilos/metabolismo , Receptores de Quimiocinas/metabolismo , Animais , Diferenciação Celular/genética , Linhagem Celular Tumoral , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/metabolismo , Metástase Neoplásica , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Receptores de Quimiocinas/genética
16.
J Transl Med ; 5: 35, 2007 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-17626627

RESUMO

BACKGROUND: Tumor cell contamination of the apheresis in multiple myeloma is likely to affect disease-free and overall survival after autografting. OBJECTIVE: To purge myeloma aphereses from tumor contaminants with a novel culture-based purging method. METHODS: We cultured myeloma-positive CD34+ PB samples in conditions that retained multipotency of hematopoietic stem cells, but were unfavourable to survival of plasma cells. Moreover, we exploited the resistance of myeloma plasma cells to retroviral transduction by targeting the hematopoietic CD34+ cell population with a retroviral vector carrying a selectable marker (the truncated form of the human receptor for nerve growth factor, DeltaNGFR). We performed therefore a further myeloma purging step by selecting the transduced cells at the end of the culture. RESULTS: Overall recovery of CD34+ cells after culture was 128.5%; DeltaNGFR transduction rate was 28.8% for CD34+ cells and 0% for CD138-selected primary myeloma cells, respectively. Recovery of CD34+ cells after DeltaNGFR selection was 22.3%. By patient-specific Ig-gene rearrangements, we assessed a decrease of 0.7-1.4 logs in tumor load after the CD34+ cell selection, and up to 2.3 logs after culture and DeltaNGFR selection. CONCLUSION: We conclude that ex-vivo culture and retroviral-mediated transduction of myeloma leukaphereses provide an efficient tumor cell purging.


Assuntos
Antígenos CD34/metabolismo , Remoção de Componentes Sanguíneos/métodos , Técnicas de Cultura de Células/métodos , Mobilização de Células-Tronco Hematopoéticas , Mieloma Múltiplo/patologia , Retroviridae/genética , Transdução Genética , Divisão Celular , Humanos , Plasmócitos/patologia , Carga Tumoral
18.
Oncotarget ; 7(38): 60793-60806, 2016 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-27563823

RESUMO

Cell fusion between neoplastic and normal cells has been suggested to play a role in the acquisition of a malignant phenotype. Several studies have pointed to the macrophage as the normal partner in this fusion, suggesting that the fused cells could acquire new invasive properties and become able to disseminate to distant organs. However, this conclusion is mainly based on studies with transplantable cell lines. We tested the occurrence of cell fusion in the MMTV-neu model of mouse mammary carcinoma. In the first approach, we generated aggregation chimeras between GFP/neu and RFP/neu embryos. Tumor cells would display both fluorescent proteins only if cell fusion with normal cells occurred. In addition, if cell fusion conferred a growth/dissemination advantage, cells with both markers should be detectable in lung metastases at increased frequency. We confirmed that fused cells are present at low but consistent levels in primary neoplasms and that the macrophage is the normal partner in the fusion events. Similar results were obtained using a second approach in which bone marrow from mice carrying the Cre transgene was transplanted into MMTV-neu/LoxP-tdTomato transgenic animals, in which the Tomato gene is activated only in the presence of CRE recombinase. However, no fused cells were detected in lung metastases in either model. We conclude that fusion between macrophages and tumor cells does not confer a selective advantage in our spontaneous model of breast cancer, although these data do not rule out a possible role in models in which an inflammation environment is prominent.


Assuntos
Macrófagos/metabolismo , Neoplasias Mamárias Animais/metabolismo , Receptor ErbB-2/metabolismo , Animais , Apoptose , Separação Celular , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Genes Reporter , Proteínas de Fluorescência Verde/metabolismo , Inflamação , Integrases/metabolismo , Masculino , Neoplasias Mamárias Experimentais/metabolismo , Vírus do Tumor Mamário do Camundongo , Camundongos , Metástase Neoplásica , Fagocitose , Fenótipo , Transgenes
19.
Mech Dev ; 116(1-2): 157-60, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12128215

RESUMO

A novel type of DNA-binding domain, the 'T-box' domain, characterizes an increasingly large family of transcription factors (Trends Genet. 15 (1999) 154). We have identified and characterized the expression pattern of a new member of the Tbr1 subfamily of T-box genes; this gene has been recently named T-bet/Tbx21 (Genomics 70 (2000) 41; Cell 17 (2000) 655; Science 292 (2001) 1907; Science 295 (2002) 338). The sequence and expression of Tbr1 and eomesodermin/Tbr2 are closely related to T-bet/Tbx21. The expression of Tbr1 (Neuron 15 (1995) 63) and Tbr2 (Mech Dev 84 (1999) 133) have virtually identical onset, at around E10.5, and expression domains in the mouse telencephalon. While Tbr1 is expressed in postmitotic neurons, Tbr2 (which is also expressed during gastrulation is also expressed in neural progenitors. We have used in situ hybridization to determine the temporal and spatial distribution of T-bet/Tbx21 expression during mouse development. T-bet/Tbx21 expression is exclusively restricted to the olfactory bulb and the thymus. To assess the distribution of T-BET/TBX21 expression in the haematopoietic compartment we used reverse transcriptase-polymerase chain reaction and found its expression in several human blood cell lineages, including progenitors/stem cells, immature B cells and peripheral T cells.


Assuntos
Proteínas com Domínio T/genética , Fatores de Transcrição/genética , Sequência de Aminoácidos , Animais , Regulação da Expressão Gênica no Desenvolvimento , Hematopoese/genética , Humanos , Hibridização In Situ , Camundongos , Dados de Sequência Molecular , RNA/genética , RNA/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos
20.
Stem Cell Reports ; 5(4): 558-68, 2015 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-26344905

RESUMO

Autosomal recessive osteopetrosis is a human bone disease mainly caused by TCIRG1 gene mutations that prevent osteoclasts resorbing activity, recapitulated by the oc/oc mouse model. Bone marrow transplantation is the only available treatment, limited by the need for a matched donor. The use of induced pluripotent stem cells (iPSCs) as an unlimited source of autologous cells to generate gene corrected osteoclasts might represent a powerful alternative. We generated iPSCs from oc/oc mice, corrected the mutation using a BAC carrying the entire Tcirg1 gene locus as a template for homologous recombination, and induced hematopoietic differentiation. Similarly to physiologic fetal hematopoiesis, iPSC-derived CD41(+) cells gradually gave rise to CD45(+) cells, which comprised both mature myeloid cells and high proliferative potential colony-forming cells. Finally, we differentiated the gene corrected iPSC-derived myeloid cells into osteoclasts with rescued bone resorbing activity. These results are promising for a future translation into the human clinical setting.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Osteoclastos/citologia , Osteopetrose/terapia , Reparo Gênico Alvo-Dirigido/métodos , ATPases Vacuolares Próton-Translocadoras/genética , Animais , Diferenciação Celular , Linhagem Celular , Hematopoese , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Células Mieloides/citologia , Células Mieloides/metabolismo , Osteoclastos/metabolismo , Osteopetrose/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA