Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Biofabrication ; 14(3)2022 05 17.
Article in English | MEDLINE | ID: mdl-35472717

ABSTRACT

Leukemia patients undergo chemotherapy to combat the leukemic cells (LCs) in the bone marrow. During therapy not only the LCs, but also the blood-producing hematopoietic stem and progenitor cells (HSPCs) may be destroyed. Chemotherapeutics targeting only the LCs are urgently needed to overcome this problem and minimize life-threatening side-effects. Predictivein vitrodrug testing systems allowing simultaneous comparison of various experimental settings would enhance the efficiency of drug development. Here, we present a three-dimensional (3D) human leukemic bone marrow model perfused using a magnetic, parallelized culture system to ensure media exchange. Chemotherapeutic treatment of the acute myeloid leukemia cell line KG-1a in 3D magnetic hydrogels seeded with mesenchymal stem/stromal cells (MSCs) revealed a greater resistance of KG-1a compared to 2D culture. In 3D tricultures with HSPCs, MSCs and KG-1a, imitating leukemic bone marrow, HSPC proliferation decreased while KG-1a cells remained unaffected post treatment. Non-invasive metabolic profiling enabled continuous monitoring of the system. Our results highlight the importance of using biomimetic 3D platforms with proper media exchange and co-cultures for creatingin vivo-like conditions to enablein vitrodrug testing. This system is a step towards drug testing in biomimetic, parallelizedin vitroapproaches, facilitating the discovery of new anti-leukemic drugs.


Subject(s)
Leukemia, Myeloid, Acute , Mesenchymal Stem Cells , Bone Marrow Cells , Cell Differentiation , Cell Proliferation , Coculture Techniques , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism
2.
Acta Biomater ; 111: 254-266, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32434077

ABSTRACT

Multiple particle tracking (MPT) microrheology was employed for monitoring the development of extracellular matrix (ECM) mechanical properties in the direct microenvironment of living cells. A customized setup enabled us to overcome current limitations: (i) Continuous measurements were enabled using a cell culture chamber, with this, matrix remodeling by fibroblasts in the heterogeneous environment of macroporous scaffolds was monitored continuously. (ii) Employing tracer laden porous scaffolds for seeding human mesenchymal stem cells (hMSCs), we followed conventional differentiation protocols. Thus, we were, for the first time able to study the massive alterations in ECM elasticity during hMSC differentiation. (iii) MPT measurements in 2D cell cultures were enabled using a long distance objective. Exemplarily, local mechanical properties of the ECM in human umbilical vein endothelial cell (HUVEC) cultures, that naturally form 2D layers, were investigated scaffold-free. Using our advanced setup, we measured local, apparent elastic moduli G0,app in a range between 0.08 and 60 Pa. For fibroblasts grown in collagen-based scaffolds, a continuous decrease of local matrix elasticity resulted during the first 10 hours after seeding. The osteogenic differentiation of hMSC cells cultivated in similar scaffolds, led to an increase of G0,app by 100 %, whereas after adipogenic differentiation it was reduced by 80 %. The local elasticity of ECM that was newly secreted by HUVECs increased significantly upon addition of protease inhibitor and in high glucose conditions even a twofold increase in G0,app was observed. The combination of these advanced methods opens up new avenues for a broad range of investigations regarding cell-matrix interactions and the propagation of ECM mechanical properties in complex biological systems. STATEMENT OF SIGNIFICANCE: Cells sense the elasticity of their environment on a micrometer length scale. For studying the local elasticity of extracellular matrix (ECM) in the direct environment of living cells, we employed an advanced multipleparticle tracking microrheology setup. MPT is based on monitoring the Brownian motion oftracer particles, which is restricted by the surrounding network. Network elasticity can thusbe quantified. Overcoming current limitations, we realized continuous investigations of ECM elasticityduring fibroblast growth. Furthermore, MPT measurements of stem cell ECM showed ECMstiffening during osteogenic differentiation and softening during adipogenic differentiation.Finally, we characterized small amounts of delicate ECM newly secreted in scaffold-freecultures of endothelial cells, that naturally form 2D layers.


Subject(s)
Mesenchymal Stem Cells , Osteogenesis , Cell Differentiation , Cellular Microenvironment , Extracellular Matrix , Humans , Tissue Scaffolds
3.
ACS Appl Bio Mater ; 3(8): 4766-4778, 2020 Aug 17.
Article in English | MEDLINE | ID: mdl-35021724

ABSTRACT

Growing numbers of complex surgical interventions increase the need for blood transfusions, which cannot be fulfilled by the number of donors. Therefore, the interest in producing erythrocytes from their precursors-the hematopoietic stem and progenitor cells (HSPCs)-in laboratories is rising. To enable this, in vitro systems are needed, which allow analysis of the effects of essential factors such as iron on erythroid development. For this purpose, iron ion-releasing systems based on poly(ethylene glycol) (PEG)-iron nanocomposites are developed to assess if gradual iron release improves iron bioavailability during in vitro erythroid differentiation. The nanocomposites are synthesized using surfactant-free pulsed laser ablation of iron directly in the PEG solution. The iron concentrations released from the material are sufficient to influence in vitro erythropoiesis. In this way, the production of erythroid cells cultured on flat PEG-iron nanocomposite hydrogel pads can be enhanced. In contrast, erythroid differentiation is not enhanced in the biomimetic macroporous 3D composite scaffolds, possibly because of local iron overload within the pores of the system. In conclusion, the developed iron nanoparticle-PEG composite hydrogel allows constant iron ion release and thus paves the way (i) to understand the role of iron during erythropoiesis and (ii) toward the development of biomaterials with a controlled iron release for directing erythropoiesis in culture.

4.
Sci Rep ; 9(1): 20003, 2019 12 27.
Article in English | MEDLINE | ID: mdl-31882795

ABSTRACT

Large or complex bone fractures often need clinical treatments for sufficient bone repair. New treatment strategies have pursued the idea of using mesenchymal stromal cells (MSCs) in combination with osteoinductive materials to guide differentiation of MSCs into bone cells ensuring complete bone regeneration. To overcome the challenge of developing such materials, fundamental studies are needed to analyze and understand the MSC behavior on modified surfaces of applicable materials for bone healing. For this purpose, we developed a fibrous scaffold resembling the bone/bone marrow extracellular matrix (ECM) based on protein without addition of synthetic polymers. With this biomimetic in vitro model we identified the fibrous structure as well as the charge of the material to be responsible for its effects on MSC differentiation. Positive charge was introduced via cationization that additionally supported the stability of the scaffold in cell culture, and acted as nucleation point for mineralization during osteogenesis. Furthermore, we revealed enhanced focal adhesion formation and osteogenic differentiation of MSCs cultured on positively charged protein fibers. This pure protein-based and chemically modifiable, fibrous ECM model allows the investigation of MSC behavior on biomimetic materials to unfold new vistas how to direct cells' differentiation for the development of new bone regenerating strategies.


Subject(s)
Cell Differentiation , Electrochemistry/methods , Mesenchymal Stem Cells/cytology , Osteogenesis , Serum Albumin, Bovine/chemistry , Biomimetic Materials , Bone Regeneration , Cations , Humans , Models, Biological , Proteolysis , Static Electricity , Surface Properties
5.
Methods Mol Biol ; 2017: 97-107, 2019.
Article in English | MEDLINE | ID: mdl-31197771

ABSTRACT

In leukemia, leukemic cells hijack the hematopoietic stem cell (HSC) microenvironment in the bone marrow-the so-called stem cell niche-by flooding the niche with clonal progeny of leukemic cells. They can exploit signaling pathways which are critical for HSC development to support their own survival, homing, and maintenance. These interactions of leukemic cells with the microenvironment have an impact on therapy progress and patient outcome. Therefore, signals for homing and anchorage of leukemic cells to the bone marrow have to be investigated by using tools that allow the migration of cells toward critical signals. Here, we describe an in vitro migration assay for leukemic cells toward a chemoattractant in a 3D environment exemplified by migration of the cell line OCI-AML3 to a CXC motif chemokine ligand 12 (CXCL12) gradient. For this purpose, a chemotaxis slide is filled with a hydrogel system mimicking the extracellular matrix in vivo. The cells are encapsulated into the hydrogel network during polymerization, and a CXCL12 gradient is introduced in the enclosed chambers to trigger migration. Cell migration in the 3D network of the hydrogel is monitored by time-lapse microscopy. We describe the experimental setup and the tools for cell tracking and data analysis.


Subject(s)
Cell Line, Tumor/cytology , Chemokine CXCL12/metabolism , Leukemia/pathology , Cell Line, Tumor/metabolism , Cell Movement , Cell Tracking , Chemotaxis , Humans , Hydrogels/chemistry , Stem Cell Niche , Tumor Microenvironment
6.
MRS Commun ; 9(1): 37-52, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30931174

ABSTRACT

The complex interaction between hematopoietic stem cells (HSCs) and their microenvironment in the human bone marrow ensures a life-long blood production by balancing stem cell maintenance and differentiation. This so-called HSC niche can be disturbed by malignant diseases. Investigating their consequences on hematopoiesis requires deep understanding of how the niches function in health and disease. To facilitate this, biomimetic models of the bone marrow are needed to analyse HSC maintenance and hematopoiesis under steady-state and diseased conditions. Here, 3D bone marrow models, their fabrication methods (including 3D bioprinting) and implementations recapturing bone marrow functions in health and diseases, are presented.

7.
Life Sci Alliance ; 1(6): e201800153, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30582132

ABSTRACT

De novo DNA methyltransferase 3A (DNMT3A) plays pivotal roles in hematopoietic differentiation. In this study, we followed the hypothesis that alternative splicing of DNMT3A has characteristic epigenetic and functional sequels. Specific DNMT3A transcripts were either down-regulated or overexpressed in human hematopoietic stem and progenitor cells, and this resulted in complementary and transcript-specific DNA methylation and gene expression changes. Functional analysis indicated that, particularly, transcript 2 (coding for DNMT3A2) activates proliferation and induces loss of a primitive immunophenotype, whereas transcript 4 interferes with colony formation of the erythroid lineage. Notably, in acute myeloid leukemia expression of transcript 2 correlates with its in vitro DNA methylation and gene expression signatures and is associated with overall survival, indicating that DNMT3A variants also affect malignancies. Our results demonstrate that specific DNMT3A variants have a distinct epigenetic and functional impact. Particularly, DNMT3A2 triggers hematopoietic differentiation and the corresponding signatures are reflected in acute myeloid leukemia.

8.
Acta Biomater ; 73: 250-262, 2018 06.
Article in English | MEDLINE | ID: mdl-29679779

ABSTRACT

In this work, we define the requirements for a human cell-based osteomyelitis model which overcomes the limitations of state of the art animal models. Osteomyelitis is a severe and difficult to treat infection of the bone that develops rapidly, making it difficult to study in humans. We have developed a 3D in vitro model of the bone marrow, comprising a macroporous material, human hematopoietic stem and progenitor cells (HSPCs) and mesenchymal stromal cells (MSCs). Inclusion of biofilms grown on an implant into the model system allowed us to study the effects of postoperative osteomyelitis-inducing bacteria on the bone marrow. The bacteria influenced the myeloid differentiation of HSPCs as well as MSC cytokine expression and the MSC ability to support HSPC maintenance. In conclusion, we provide a new 3D in vitro model which meets all the requirements for investigating the impact of osteomyelitis. STATEMENT OF SIGNIFICANCE: Implant-associated osteomyelitis is a persistent bacterial infection of the bone which occurs in many implant patients and can result in functional impairments or even entire loss of the extremity. Nevertheless, surprisingly little is known on the triangle interaction between implant material, bacterial biofilm and affected bone tissue. Closing this gap of knowledge would be crucial for the fundamental understanding of the disease and the development of novel treatment strategies. For this purpose, we developed the first biomaterial-based system that is able to mimic implant-associated osteomyelitis outside of the body, thus, opening the avenue to study this fatal disease in the laboratory.


Subject(s)
Biofilms/growth & development , Biomimetic Materials/pharmacology , Bone Marrow Diseases , Hematopoiesis , Implants, Experimental/microbiology , Methicillin-Resistant Staphylococcus aureus/physiology , Models, Biological , Osteomyelitis , Staphylococcal Infections , Bone Marrow Diseases/metabolism , Bone Marrow Diseases/microbiology , Bone Marrow Diseases/pathology , Cells, Cultured , Humans , Implants, Experimental/adverse effects , Osteomyelitis/metabolism , Osteomyelitis/microbiology , Osteomyelitis/pathology , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology
9.
Adv Healthc Mater ; 7(9): e1701403, 2018 05.
Article in English | MEDLINE | ID: mdl-29349923

ABSTRACT

There is an urgent need for 3D cell culture systems that avoid the oversimplifications and artifacts of conventional culture in 2D. However, 3D culture within the cavities of porous biomaterials or large 3D structures harboring high cell numbers is limited by the needs to nurture cells and to remove growth-limiting metabolites. To overcome the diffusion-limited transport of such soluble factors in 3D culture, mixing can be improved by pumping, stirring or shaking, but this in turn can lead to other problems. Using pumps typically requires custom-made accessories that are not compatible with conventional cell culture disposables, thus interfering with cell production processes. Stirring or shaking allows little control over movement of scaffolds in media. To overcome these limitations, magnetic, macroporous hydrogels that can be moved or positioned within media in conventional cell culture tubes in a contactless manner are presented. The cytocompatibility of the developed biomaterial and the applied magnetic fields are verified for human hematopoietic stem and progenitor cells (HSPCs). The potential of this technique for perfusing 3D cultures is demonstrated in a proof-of-principle study that shows that controlled contactless movement of cell-laden magnetic hydrogels in culture media can mimic the natural influence of differently perfused environments on HSPCs.


Subject(s)
Cell Culture Techniques/methods , Hematopoietic Stem Cells/metabolism , Hydrogels/chemistry , Magnetic Fields , Tissue Scaffolds/chemistry , Hematopoietic Stem Cells/cytology , Humans , Porosity
10.
Methods Mol Biol ; 1202: 121-30, 2014.
Article in English | MEDLINE | ID: mdl-24875249

ABSTRACT

In vitro proliferation of hematopoietic stem cells (HSCs) is yet an unresolved challenge. Found in the bone marrow, HSCs can undergo self-renewing cell division and thereby multiply. Recapitulation of the bone marrow environment in order to provide the required signals for their expansion is a promising approach.Here, we describe a technique to produce biofunctionalized, macroporous poly(ethylene glycol) diacrylate (PEGDA) hydrogels that mimic the spongy 3D architecture of trabecular bones, which host the red, blood-forming bone marrow. After seeding these scaffolds with cells, they can be used as simplified bone marrow analogs for the cultivation of HSCs. This method can easily be conducted with standard laboratory chemicals and equipment. The 3D hydrogels are produced via salt leaching and biofunctionalization of the material is achieved by co-polymerizing the PEGDA with an RGD peptide. Finally, cell seeding and retrieval are described.


Subject(s)
Bone Marrow/physiology , Cell Culture Techniques/methods , Hematopoietic Stem Cells/cytology , Hydrogel, Polyethylene Glycol Dimethacrylate/pharmacology , Polyethylene Glycols/pharmacology , Bone Marrow/drug effects , Cells, Cultured , Hematopoietic Stem Cells/drug effects , Humans , Porosity
11.
Biomaterials ; 35(3): 929-40, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24176196

ABSTRACT

Multiplication of hematopoietic stem cells (HSCs) in vitro with current standard methods is limited and mostly insufficient for clinical applications of these cells. They quickly lose their multipotency in culture because of the fast onset of differentiation. In contrast, HSCs efficiently self-renew in their natural microenvironment (their niche) in the bone marrow. Therefore, engineering artificial bone marrow analogs is a promising biomaterial-based approach for culturing these cells. In the current study, a straight-forward, easy-to-use method for the production of biofunctionalized, macroporous hydrogel scaffolds that mimic the spongy architecture of trabecular bones was developed. As surrogates for cellular components of the niche, mesenchymal stem cells (MSCs) from different sources (bone marrow and umbilical cord) and osteoblast-like cells were tested. MSCs from bone marrow had the strongest pro-proliferative effect on freshly isolated human hematopoietic stem and progenitor cells (HSPCs) from umbilical cord blood. Co-culture in the pores of the three-dimensional hydrogel scaffold showed that the positive effect of MSCs on preservation of HSPC stemness was more pronounced in 3D than in standard 2D cell culture systems. Thus, the presented biomimetic scaffolds revealed to meet the basic requirements for creating artificial HSC niches.


Subject(s)
Hematopoietic Stem Cells/cytology , Hydrogels/chemistry , Polyethylene Glycols/chemistry , Tissue Scaffolds/chemistry , Biomimetic Materials/chemistry , Biomimetics , Bone Marrow Cells/cytology , Cells, Cultured , Coculture Techniques , Fetal Blood/cytology , Humans , Mesenchymal Stem Cells/cytology , Porosity
SELECTION OF CITATIONS
SEARCH DETAIL
...