Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Stem Cell Res Ther ; 15(1): 124, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38679735

ABSTRACT

BACKGROUND: Recombinant human bone morphogenetic protein 2 (rhBMP-2) and human bone marrow mesenchymal stromal cells (hBM-MSCs) have been thoroughly studied for research and translational bone regeneration purposes. rhBMP-2 induces bone formation in vivo, and hBM-MSCs are its target, bone-forming cells. In this article, we studied how rhBMP-2 drives the multilineage differentiation of hBM-MSCs both in vivo and in vitro. METHODS: rhBMP-2 and hBM-MSCs were tested in an in vivo subcutaneous implantation model to assess their ability to form mature bone and undergo multilineage differentiation. Then, the hBM-MSCs were treated in vitro with rhBMP-2 for short-term or long-term cell-culture periods, alone or in combination with osteogenic, adipogenic or chondrogenic media, aiming to determine the role of rhBMP-2 in these differentiation processes. RESULTS: The data indicate that hBM-MSCs respond to rhBMP-2 in the short term but fail to differentiate in long-term culture conditions; these cells overexpress the rhBMP-2 target genes DKK1, HEY-1 and SOST osteogenesis inhibitors. However, in combination with other differentiation signals, rhBMP-2 acts as a potentiator of multilineage differentiation, not only of osteogenesis but also of adipogenesis and chondrogenesis, both in vitro and in vivo. CONCLUSIONS: Altogether, our data indicate that rhBMP-2 alone is unable to induce in vitro osteogenic terminal differentiation of hBM-MSCs, but synergizes with other signals to potentiate multiple differentiation phenotypes. Therefore, rhBMP-2 triggers on hBM-MSCs different specific phenotype differentiation depending on the signalling environment.


Subject(s)
Bone Morphogenetic Protein 2 , Cell Differentiation , Mesenchymal Stem Cells , Osteogenesis , Recombinant Proteins , Transforming Growth Factor beta , Humans , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Bone Morphogenetic Protein 2/pharmacology , Bone Morphogenetic Protein 2/metabolism , Cell Differentiation/drug effects , Recombinant Proteins/pharmacology , Osteogenesis/drug effects , Animals , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology , Chondrogenesis/drug effects , Cells, Cultured , Mice , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Marrow Cells/drug effects , Signal Transduction/drug effects , Adipogenesis/drug effects
2.
ACS Appl Mater Interfaces ; 16(13): 15993-16002, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38509001

ABSTRACT

Biomaterials capable of delivering therapeutic proteins are relevant in biomedicine, yet their manufacturing relies on centralized manufacturing chains that pose challenges to their remote implementation at the point of care. This study explores the viability of confined cell-free protein synthesis within porous hydrogels as biomaterials that dynamically produce and deliver proteins to in vitro and in vivo biological microenvironments. These functional biomaterials have the potential to be assembled as implants at the point of care. To this aim, we first entrap cell-free extracts (CFEs) from Escherichia coli containing the transcription-translation machinery, together with plasmid DNA encoding the super folded green fluorescence protein (sGFP) as a model protein, into hydrogels using various preparation methods. Agarose hydrogels result in the most suitable biomaterials to confine the protein synthesis system, demonstrating efficient sGFP production and diffusion from the core to the surface of the hydrogel. Freeze-drying (FD) of agarose hydrogels still allows for the synthesis and diffusion of sGFP, yielding a more attractive biomaterial for its reconstitution and implementation at the point of care. FD-agarose hydrogels are biocompatible in vitro, allowing for the colonization of cell microenvironments along with cell proliferation. Implantation assays of this biomaterial in a preclinical mouse model proved the feasibility of this protein synthesis approach in an in vivo context and indicated that the physical properties of the biomaterials influence their immune responses. This work introduces a promising avenue for biomaterial fabrication, enabling the in vivo synthesis and targeted delivery of proteins and opening new paths for advanced protein therapeutic approaches based on biocompatible biomaterials.


Subject(s)
Biocompatible Materials , Hydrogels , Animals , Mice , Biocompatible Materials/pharmacology , Biocompatible Materials/therapeutic use , Hydrogels/therapeutic use , Sepharose , Prostheses and Implants
3.
Biomater Sci ; 11(15): 5163-5176, 2023 Jul 25.
Article in English | MEDLINE | ID: mdl-37435668

ABSTRACT

Digital light processing (DLP) printing offers the possibility of fabricating complex objects in a fast and reproducible manner. A main requirement for DLP printing is the use of inks with low viscosities that can flow under the printing platform in a short period of time. Its exploitation in tissue engineering applications has been centered on the use of hydrogel forming materials diluted in aqueous solutions or the use of polyesters in combination with diluents and heating platforms that aid in the reduction of their viscosity. The use of diluents, however, modifies the mechanical properties and reduces the shape fidelity of the printed objects and, the use of heating platforms results in vats with heterogeneous temperatures and ink viscosities. Here, we report on the synthesis of a library of methacrylated low molecular weight (<3000 g mol-1) homopolymers ((P(D,L)LA and PCL) and copolymers (P((D,L)LA-co-CL)) of 2- and 3-arms based on (D,L)-lactide and ε-caprolactone. The resulting inks possessed low viscosity that made them printable in the absence of diluents and heating elements. DLP printing of cubical and cylindrical patterns resulted in objects with a higher shape fidelity than their counterparts fabricated using diluents and with printed features on the order of 300 µm. The printed materials were biocompatible and supported the growth of human mesenchymal stem cells (hMSCs). Moreover, the variations in the composition resulted in polymers that enabled the attachment of hMSCs to different extents, leading to the formation of well-adhered cell monolayers or loosely adhered cell aggregates.


Subject(s)
Biocompatible Materials , Ink , Humans , Molecular Weight , Polymers , Polyesters , Tissue Engineering , Printing, Three-Dimensional , Cell Culture Techniques
4.
Biomater Adv ; 151: 213465, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37236118

ABSTRACT

Biocompatible three-dimensional porous scaffolds are widely used in multiple biomedical applications. However, the fabrication of tailor-made 3D structures with controlled and combined multiscale macroscopic-microscopic, surface and inner porosities in a straightforward manner is still a current challenge. Herein, we use multimaterial fused deposition modeling (FDM) to generate poly (vinyl alcohol) (PVA) sacrificial moulds filled with poly (Ɛ-caprolactone) (PCL) to generate well defined PCL 3D objects. Further on, the supercritical CO2 (SCCO2) technique, as well as the breath figures mechanism (BFs), were additionally employed to fabricate specific porous structures at the core and surfaces of the 3D PCL object, respectively. The biocompatibility of the resulting multiporous 3D structures was tested in vitro and in vivo, and the versatility of the approach was assessed by generating a vertebra model fully tunable at multiple pore size levels. In sum, the combinatorial strategy to generate porous scaffolds offers unique possibilities to fabricate intricate structures by combining the advantages of additive manufacturing (AM), which provides flexibility and versatility to generate large sized 3D structures, with advantages of the SCCO2 and BFs techniques, which allow to finely tune the macro and micro porosity at material surface and material core levels.


Subject(s)
Tissue Engineering , Tissue Scaffolds , Tissue Scaffolds/chemistry , Tissue Engineering/methods , Porosity , Polyvinyl Alcohol , Printing, Three-Dimensional
5.
Nat Commun ; 13(1): 7619, 2022 Dec 09.
Article in English | MEDLINE | ID: mdl-36494342

ABSTRACT

Myelodysplastic syndromes (MDS) are hematopoietic stem cell (HSC) malignancies characterized by ineffective hematopoiesis, with increased incidence in older individuals. Here we analyze the transcriptome of human HSCs purified from young and older healthy adults, as well as MDS patients, identifying transcriptional alterations following different patterns of expression. While aging-associated lesions seem to predispose HSCs to myeloid transformation, disease-specific alterations may trigger MDS development. Among MDS-specific lesions, we detect the upregulation of the transcription factor DNA Damage Inducible Transcript 3 (DDIT3). Overexpression of DDIT3 in human healthy HSCs induces an MDS-like transcriptional state, and dyserythropoiesis, an effect associated with a failure in the activation of transcriptional programs required for normal erythroid differentiation. Moreover, DDIT3 knockdown in CD34+ cells from MDS patients with anemia is able to restore erythropoiesis. These results identify DDIT3 as a driver of dyserythropoiesis, and a potential therapeutic target to restore the inefficient erythroid differentiation characterizing MDS patients.


Subject(s)
Myelodysplastic Syndromes , Transcription Factors , Adult , Humans , Aged , Transcription Factors/genetics , Transcription Factors/metabolism , Myelodysplastic Syndromes/pathology , Erythropoiesis/genetics , Hematopoietic Stem Cells/metabolism , Gene Expression Regulation , Transcription Factor CHOP/genetics
6.
Front Oncol ; 12: 956940, 2022.
Article in English | MEDLINE | ID: mdl-36059712

ABSTRACT

In vitro cell culture studies are common in the cancer research field, and reliable biomimetic 3D models are needed to ensure physiological relevance. In this manuscript, we hypothesized that decellularized xenograft tumors can serve as an optimal 3D substrate to generate a top-down approach for in vitro tumor modeling. Multiple tumor cell lines were xenografted and the formed solid tumors were recovered for their decellularization by several techniques and further characterization by histology and proteomics techniques. Selected decellularized tumor xenograft samples were seeded with the HCC1806 human triple-negative breast cancer (TNBC) basal-like subtype cell line, and cell behavior was compared among them and with other control 2D and 3D cell culture methods. A soft treatment using Freeze-EDTA-DNAse allows proper decellularization of xenografted tumor samples. Interestingly, proteomic data show that samples decellularized from TNBC basal-like subtype xenograft models had different extracellular matrix (ECM) compositions compared to the rest of the xenograft tumors tested. The in vitro recellularization of decellularized ECM (dECM) yields tumor-type-specific cell behavior in the TNBC context. Data show that dECM derived from xenograft tumors is a feasible substrate for reseeding purposes, thereby promoting tumor-type-specific cell behavior. These data serve as a proof-of-concept for further potential generation of patient-specific in vitro research models.

7.
Polymers (Basel) ; 14(12)2022 Jun 15.
Article in English | MEDLINE | ID: mdl-35745998

ABSTRACT

Biodegradable membranes, including Polylactic acid (PLA)-based membranes, are commonly used in bone-tissue-related clinical procedures as biointerface to promote bone tissue regeneration. Calcium (Ca2+) and Magnesium (Mg2+) ions have been related to the promotion of osteogenesis, where the PLA membranes could be used as carrier and delivery substrate for them to provide osteogenic properties to this material. For this aim, a new ion delivery system based on biodegradable PLA membranes loaded with Mg and hydroxyapatite (HA) particles has been processed by the combination of tape casting and colloidal route. Materials characterization shows that the incorporation of Mg and HA particles changes the surface and hydrophobicity of the PLA membrane, and the in vitro degradation test shows Mg2+ and Ca2+ ion release and occasionally the precipitation of different ion species onto the membrane surface. Mouse and human Mesenchymal Stem Cells (MSC) were used to define the biocompatibility and bioactivity of these PLA membrane composites, and data indicated Mg2+ promotes cell proliferation and potentiates osteoinductive signals, while Ca2+ induces the expression of ALP osteogenic marker in human MSCs. Biodegradable PLA membranes loaded with Mg and HA particles is a promising new ion delivery system of Mg2+ and Ca2+ ions that provides osteogenic signals and works as functional biointerface interfaces with bone tissues.

8.
Cell Rep ; 35(6): 109119, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33979628

ABSTRACT

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.


Subject(s)
Bone Marrow Cells/metabolism , Leukemia, Myeloid, Acute/genetics , Proteomics/methods , Animals , Humans , Mice , Tumor Microenvironment
9.
Blood Cancer Discov ; 2(2): 135-145, 2021 03.
Article in English | MEDLINE | ID: mdl-33778768

ABSTRACT

Myelodysplastic syndrome (MDS) are clonal stem cell diseases characterized mainly by ineffective hematopoiesis. Here, we present an approach that enables robust long-term engraftment of primary MDS stem cells (MDS-SCs) in mice by implantation of human mesenchymal cell-seeded scaffolds. Critically for modelling MDS, where patient sample material is limiting, mononuclear bone marrow cells containing as few as 104 CD34+ cells can be engrafted and expanded by this approach with the maintenance of the genetic make-up seen in the patients. Non-invasive high-resolution ultrasound imaging shows that these scaffolds are fully perfused. Our data shows that human microenvironment but not mouse is essential to MDS-SCs homing and engraftment. Notably, the alternative niche provided by healthy donor MSCs enhanced engraftment of MDS-SCs. This study characterizes a new tool to model MDS human disease with the level of engraftment previously unattainable in mice, and offers insights into human-specific determinants of MDS-SC microenvironment.


Subject(s)
Mesenchymal Stem Cells , Myelodysplastic Syndromes , Animals , Bone Marrow Cells , Hematopoiesis , Humans , Mice , Stem Cells
10.
Int J Mol Sci ; 21(15)2020 Jul 30.
Article in English | MEDLINE | ID: mdl-32751654

ABSTRACT

The extracellular matrix (ECM) is a complex network with multiple functions, including specific functions during tissue regeneration. Precisely, the properties of the ECM have been thoroughly used in tissue engineering and regenerative medicine research, aiming to restore the function of damaged or dysfunctional tissues. Tissue decellularization is gaining momentum as a technique to obtain potentially implantable decellularized extracellular matrix (dECM) with well-preserved key components. Interestingly, the tissue-specific dECM is becoming a feasible option to carry out regenerative medicine research, with multiple advantages compared to other approaches. This review provides an overview of the most common methods used to obtain the dECM and summarizes the strategies adopted to decellularize specific tissues, aiming to provide a helpful guide for future research development.


Subject(s)
Extracellular Matrix/genetics , Regenerative Medicine/trends , Tissue Engineering , Humans , Musculoskeletal System/chemistry , Musculoskeletal System/metabolism , Tissue Scaffolds/chemistry
11.
J Clin Invest ; 130(6): 3038-3050, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32364536

ABSTRACT

Acute myeloid leukemia (AML) disrupts the generation of normal blood cells, predisposing patients to hemorrhage, anemia, and infections. Differentiation and proliferation of residual normal hematopoietic stem and progenitor cells (HSPCs) are impeded in AML-infiltrated bone marrow (BM). The underlying mechanisms and interactions of residual hematopoietic stem cells (HSCs) within the leukemic niche are poorly understood, especially in the human context. To mimic AML infiltration and dissect the cellular crosstalk in human BM, we established humanized ex vivo and in vivo niche models comprising AML cells, normal HSPCs, and mesenchymal stromal cells (MSCs). Both models replicated the suppression of phenotypically defined HSPC differentiation without affecting their viability. As occurs in AML patients, the majority of HSPCs were quiescent and showed enrichment of functional HSCs. HSPC suppression was largely dependent on secreted factors produced by transcriptionally remodeled MSCs. Secretome analysis and functional validation revealed MSC-derived stanniocalcin 1 (STC1) and its transcriptional regulator HIF-1α as limiting factors for HSPC proliferation. Abrogation of either STC1 or HIF-1α alleviated HSPC suppression by AML. This study provides a humanized model to study the crosstalk among HSPCs, leukemia, and their MSC niche, and a molecular mechanism whereby AML impairs normal hematopoiesis by remodeling the mesenchymal niche.


Subject(s)
Glycoproteins/metabolism , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Leukemia, Myeloid, Acute/metabolism , Mesenchymal Stem Cells/metabolism , Neoplasm Proteins/metabolism , Animals , Female , Glycoproteins/genetics , HL-60 Cells , Hematopoietic Stem Cells/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Neoplasm Proteins/genetics , U937 Cells
12.
Blood ; 136(7): 885-897, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32294156

ABSTRACT

Idiopathic aplastic anemia (AA) has 2 key characteristics: an autoimmune response against hematopoietic stem/progenitor cells and regulatory T-cells (Tregs) deficiency. We have previously demonstrated reduction in a specific subpopulation of Treg in AA, which predicts response to immunosuppression. The aims of the present study were to define mechanisms of Treg subpopulation imbalance and identify potential for therapeutic intervention. We have identified 2 mechanisms that lead to skewed Treg composition in AA: first, FasL-mediated apoptosis on ligand interaction; and, second, relative interleukin-2 (IL-2) deprivation. We have shown that IL-2 augmentation can overcome these mechanisms. Interestingly, when high concentrations of IL-2 were used for in vitro Treg expansion cultures, AA Tregs were able to expand. The expanded populations expressed a high level of p-BCL-2, which makes them resistant to apoptosis. Using a xenograft mouse model, the function and stability of expanded AA Tregs were tested. We have shown that these Tregs were able to suppress the macroscopic clinical features and tissue manifestations of T-cell-mediated graft-versus-host disease. These Tregs maintained their suppressive properties as well as their phenotype in a highly inflammatory environment. Our findings provide an insight into the mechanisms of Treg reduction in AA. We have identified novel targets with potential for therapeutic interventions. Supplementation of ex vivo expansion cultures of Tregs with high concentrations of IL-2 or delivery of IL-2 directly to patients could improve clinical outcomes in addition to standard immunosuppressive therapy.


Subject(s)
Anemia, Aplastic/immunology , Apoptosis/drug effects , Fas Ligand Protein/pharmacology , Interleukin-2/pharmacology , T-Lymphocytes, Regulatory/drug effects , Anemia, Aplastic/pathology , Animals , Apoptosis/immunology , Cells, Cultured , Female , Humans , Immune System Diseases/immunology , Immune System Diseases/pathology , Immune Tolerance/drug effects , Immune Tolerance/immunology , Interleukin-2/deficiency , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , T-Lymphocytes, Regulatory/physiology
14.
Leukemia ; 34(6): 1658-1668, 2020 06.
Article in English | MEDLINE | ID: mdl-31776464

ABSTRACT

Juvenile myelomonocytic leukemia (JMML) is a rare aggressive myelodysplastic/myeloproliferative neoplasm of early childhood, initiated by RAS-activating mutations. Genomic analyses have recently described JMML mutational landscape; however, the nature of JMML-propagating cells (JMML-PCs) and the clonal architecture of the disease remained until now elusive. Combining genomic (exome, RNA-seq), Colony forming assay and xenograft studies, we detect the presence of JMML-PCs that faithfully reproduce JMML features including the complex/nonlinear organization of dominant/minor clones, both at diagnosis and relapse. Further integrated analysis also reveals that although the mutations are acquired in hematopoietic stem cells, JMML-PCs are not always restricted to this compartment, highlighting the heterogeneity of the disease during the initiation steps. We show that the hematopoietic stem/progenitor cell phenotype is globally maintained in JMML despite overexpression of CD90/THY-1 in a subset of patients. This study shed new lights into the ontogeny of JMML, and the identity of JMML-PCs, and provides robust models to monitor the disease and test novel therapeutic approaches.


Subject(s)
Hematopoietic Stem Cells/pathology , Leukemia, Myelomonocytic, Juvenile/pathology , Neoplastic Stem Cells/pathology , Adolescent , Animals , Child , Child, Preschool , Female , Heterografts , Humans , Infant , Leukemia, Myelomonocytic, Juvenile/genetics , Male , Mice , Mutation
15.
J Clin Med ; 8(12)2019 12 11.
Article in English | MEDLINE | ID: mdl-31835761

ABSTRACT

A cogent issue in cancer research is how to account for the effects of tumor microenvironment (TME) on the response to therapy, warranting the need to adopt adequate in vitro and in vivo models. This is particularly relevant in the development of strategies targeting cancer metabolism, as they will inevitably have systemic effects. For example, inhibition of mitochondrial complex I (CI), despite showing promising results as an anticancer approach, triggers TME-mediated survival mechanisms in subcutaneous osteosarcoma xenografts, a response that may vary according to whether the tumors are induced via subcutaneous injection or by intrabone orthotopic transplantation. Thus, with the aim to characterize the TME of CI-deficient tumors in a model that more faithfully represents osteosarcoma development, we set up a humanized bone niche ectopic graft. A prominent involvement of TME was revealed in CI-deficient tumors, characterized by the abundance of cancer associated fibroblasts, tumor associated macrophages and preservation of osteocytes and osteoblasts in the mineralized bone matrix. The pseudo-orthotopic approach allowed investigation of osteosarcoma progression in a bone-like microenvironment setting, without being invasive as the intrabone cell transplantation. Additionally, establishing osteosarcomas in a humanized bone niche model identified a peculiar association between targeting CI and bone tissue preservation.

16.
Sci Rep ; 8(1): 15615, 2018 10 23.
Article in English | MEDLINE | ID: mdl-30353072

ABSTRACT

Mesenchymal progenitor cells (MPCs) have been hypothesized as cells of origin for sarcomas, and c-Fos transcription factor has been showed to act as an oncogene in bone tumors. In this study, we show c-Fos is present in most sarcomas with chondral phenotype, while multiple other genes are related to c-Fos expression pattern. To further define the role of c-Fos in sarcomagenesis, we expressed it in primary human MPCs (hMPCs), immortalized hMPCs and transformed murine MPCs (mMPCs). In immortalized hMPCs, c-Fos expression generated morphological changes, reduced mobility capacity and impaired adipogenic- and osteogenic-differentiation potentials. Remarkably, immortalized hMPCs or mMPCs expressing c-Fos generated tumors harboring a chondrogenic phenotype and morphology. Thus, here we show that c-Fos protein has a key role in sarcomas and that c-Fos expression in immortalized MPCs yields cell transformation and chondrogenic tumor formation.


Subject(s)
Carcinogenesis/genetics , Cell Transformation, Neoplastic/genetics , Mesenchymal Stem Cells/pathology , Proto-Oncogene Proteins c-fos/genetics , Sarcoma/genetics , Animals , Carcinogenesis/pathology , Cell Line , Cell Transformation, Neoplastic/pathology , Gene Expression Regulation, Neoplastic , Genes, fos , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Proto-Oncogene Proteins c-fos/analysis , Sarcoma/pathology
17.
Nat Commun ; 9(1): 3994, 2018 09 28.
Article in English | MEDLINE | ID: mdl-30266933

ABSTRACT

Osteosarcoma is a type of bone tumour characterized by considerable levels of phenotypic heterogeneity, aneuploidy, and a high mutational rate. The life expectancy of osteosarcoma patients has not changed during the last three decades and thus much remains to be learned about the disease biology. Here, we employ a RGB-based single-cell tracking system to study the clonal dynamics occurring in a de novo-induced murine osteosarcoma model. We show that osteosarcoma cells present initial polyclonal dynamics, followed by clonal dominance associated with adaptation to the microenvironment. Interestingly, the dominant clones are composed of subclones with a similar tumour generation potential when they are re-implanted in mice. Moreover, individual spontaneous metastases are clonal or oligoclonal, but they have a different cellular origin than the dominant clones present in primary tumours. In summary, we present evidence that osteosarcomagenesis can follow a neutral evolution model, in which different cancer clones coexist and propagate simultaneously.


Subject(s)
Bone Neoplasms/metabolism , Clone Cells/metabolism , Luminescent Proteins/metabolism , Osteosarcoma/metabolism , Animals , Bone Neoplasms/genetics , Luminescent Proteins/genetics , Mesenchymal Stem Cells/metabolism , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , Microscopy, Confocal , Osteosarcoma/genetics , Single-Cell Analysis/methods
18.
Stem Cells ; 36(10): 1487-1500, 2018 10.
Article in English | MEDLINE | ID: mdl-30001480

ABSTRACT

Osteosarcoma (OS) is a highly aggressive bone tumor that usually arises intramedullary at the extremities of long bones. Due to the fact that the peak of incidence is in the growth spurt of adolescence, the specific anatomical location, and the heterogeneity of cells, it is believed that osteosarcomagenesis is a process associated with bone development. Different studies in murine models showed that the tumor-initiating cell in OS could be an uncommitted mesenchymal stem cell (MSC) developing in a specific bone microenvironment. However, only a few studies have reported transgene-induced human MSCs transformation and mostly obtained undifferentiated sarcomas. In our study, we demonstrate that activator protein 1 family members induce osteosarcomagenesis in immortalized hMSC. c-JUN or c-JUN/c-FOS overexpression act as tumorigenic factors generating OS with fibroblastic or pleomorphic osteoblastic phenotypes, respectively. Stem Cells 2018;36:1487-1500.


Subject(s)
Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Osteosarcoma/metabolism , Osteosarcoma/pathology , Transcription Factor AP-1/metabolism , Animals , Heterografts , Humans , Mice , Mice, SCID , Phenotype
19.
J Exp Med ; 215(3): 729-743, 2018 03 05.
Article in English | MEDLINE | ID: mdl-29453226

ABSTRACT

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.


Subject(s)
Bioengineering/methods , Bone Marrow Transplantation , Bone Marrow/metabolism , Stem Cell Niche , Animals , Hematopoiesis , Humans , Mice , Transplantation, Heterologous
20.
Nat Commun ; 8(1): 1679, 2017 11 22.
Article in English | MEDLINE | ID: mdl-29162833

ABSTRACT

Cytarabine (AraC) represents the most effective single agent treatment for AML. Nevertheless, overriding AraC resistance in AML remains an unmet medical need. Here we show that the CHK1 inhibitor (CHK1i) GDC-0575 enhances AraC-mediated killing of AML cells both in vitro and in vivo, thus abrogating any potential chemoresistance mechanisms involving DNA repair. Importantly, this combination of drugs does not affect normal long-term hematopoietic stem/progenitors. Moreover, the addition of CHK1i to AraC does not generate de novo mutations and in patients' samples where AraC is mutagenic, addition of CHK1i appears to eliminate the generation of mutant clones. Finally, we observe that persistent residual leukemic cells are quiescent and can become responsive to the treatment when forced into cycle via granulocyte colony-stimulating factor (G-CSF) administration. This drug combination (AraC+CHK1i+G-CSF) will open the doors for a more efficient treatment of AML in the clinic.


Subject(s)
Checkpoint Kinase 1/antagonists & inhibitors , Cytarabine/administration & dosage , Granulocyte Colony-Stimulating Factor/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Piperidines/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Pyridines/administration & dosage , Pyrroles/administration & dosage , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , HL-60 Cells , Hematopoiesis/drug effects , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mutation/drug effects , U937 Cells , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...