Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Hematology ; 27(1): 476-487, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35413231

ABSTRACT

OBJECTIVE: The interplay between intrinsic and extrinsic elements involved in the physiology of hematopoietic cells is not completely understood. In the present study, we analyzed the transcriptional profiles of human cord blood-derived hematopoietic stem cells (HSCs), as well as myeloid (MPCs) and erythroid (EPCs) progenitors, and assessed their proliferation and expansion kinetics in vitro. METHODS: All cell populations were obtained by cell-sorting, and were cultured in liquid cultures supplemented with different cytokine combinations. Their gene expression profiles were determined by RNA microarrays right after cell-sorting, before culture. RESULTS: HSCs showed the highest proliferation and expansion capacities in culture, and were found to be more closely related, in transcriptional terms, to MPCs than to EPCs. This correlated with the fact that after 30 days, only cultures initiated with HSCs and MPCs were sustained. Expression of cell cycle and cell division-related genes was enriched in EPCs. Such cells showed significantly higher proliferation than MPCs, however, their expansion potential was reduced, so that cultures initiated with EPCs declined after 15 days and became exhausted by day 30. Proliferation and expansion of HSCs and EPCs were higher in the presence of a cytokine combination that favors erythropoiesis, whereas the growth of MPCs was higher under a cytokine combination that favors myelopoiesis. CONCLUSION: This study shows a correlation between the transcriptional profiles of HSCs, MPCs, and EPCs, and their respective in vitro growth under particular culture conditions. These results may be relevant in the development of ex vivo systems for the expansion of hematopoietic cells for clinical application.


Subject(s)
Cytokines , Hematopoietic Stem Cells , Antigens, CD34/metabolism , Cell Proliferation , Cells, Cultured , Cytokines/genetics , Fetal Blood/metabolism , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/physiology , Humans , Transcriptome
2.
STAR Protoc ; 2(4): 100846, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34622219

ABSTRACT

Hematopoietic stem and progenitor cells (HSPCs) reside in the bone marrow and supply blood cells. Efficient methods for isolation of HSPCs are required. Here, we present protocols for the isolation of human and murine HSPCs using manual and FACS-assisted techniques. Isolated HSPCs can be used for downstream applications, including colony forming unit assays and DNA damage and repair assays. For complete details on the use and execution of this protocol, please refer to Rodríguez et al. (2021a) and (2021b).


Subject(s)
Bone Marrow , Hematopoietic Stem Cells , Animals , Colony-Forming Units Assay , DNA Damage/genetics , DNA Repair , Humans , Mice
3.
Exp Hematol ; 93: 70-84.e4, 2021 01.
Article in English | MEDLINE | ID: mdl-33166613

ABSTRACT

Fanconi anemia (FA) is a chromosome instability syndrome with congenital abnormalities, cancer predisposition and bone marrow failure (BMF). Although hematopoietic stem and progenitor cell (HSPC) transplantation is the recommended therapy, new therapies are needed for FA patients without suitable donors. BMF in FA is caused, at least in part, by a hyperactive growth-suppressive transforming growth factor ß (TGFß) pathway, regulated by the TGFß1, TGFß2, and TGFß3 ligands. Accordingly, the TGFß pathway is an attractive therapeutic target for FA. While inhibition of TGFß1 and TGFß3 promotes blood cell expansion, inhibition of TGFß2 is known to suppress hematopoiesis. Here, we report the effects of AVID200, a potent TGFß1- and TGFß3-specific inhibitor, on FA hematopoiesis. AVID200 promoted the survival of murine FA HSPCs in vitro. AVID200 also promoted in vitro the survival of human HSPCs from patients with FA, with the strongest effect in patients progressing to severe aplastic anemia or myelodysplastic syndrome (MDS). Previous studies have indicated that the toxic upregulation of the nonhomologous end-joining (NHEJ) pathway accounts, at least in part, for the poor growth of FA HSPCs. AVID200 downregulated the expression of NHEJ-related genes and reduced DNA damage in primary FA HSPC in vitro and in in vivo models. Collectively, AVID200 exhibits activity in FA mouse and human preclinical models. AVID200 may therefore provide a therapeutic approach to improving BMF in FA.


Subject(s)
Fanconi Anemia/drug therapy , Hematopoiesis/drug effects , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta3/antagonists & inhibitors , Adolescent , Adult , Animals , Cell Survival/drug effects , Cells, Cultured , Child , Child, Preschool , Fanconi Anemia/metabolism , Fanconi Anemia/physiopathology , Female , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/pathology , Humans , Male , Mice , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta3/metabolism
4.
Blood Cells Mol Dis ; 85: 102485, 2020 11.
Article in English | MEDLINE | ID: mdl-32836190

ABSTRACT

In vitro growth of hematopoietic cells depends on the presence of hematopoietic cytokines. To date, it is unclear if these cells would be able to respond to non-hematopoietic cytokines. In the present study, we have explored this by culturing human hematopoietic cells in presence of neurogenic cytokines. Lineage-negative (Lin-) umbilical cord blood (UCB)-derived cells -enriched for hematopoietic stem and progenitor cells- were cultured in presence of different combinations of hematopoietic cytokines, neurotrophins, epidermal growth factor, fibroblast growth factor, and neurogenic culture media, in a 3-phase culture system. A proportion (1-22%) of Lin- UCB hematopoietic cells normally express neural markers and are capable of responding to neural cytokines. Neural cytokines did not have effects on hematopoietic cell proliferation; however, we observed generation of neural-like cells, assessed by morphology, and a significant increase in the proportion of cells expressing neural markers. Such neural-like cells, however, retained expression of hematopoietic markers. It seems that under our culture conditions, no actual transdifferentiation of hematopoietic cells into neural cells occurred; instead, the cells generated in culture seem to be hematopoietic cells that acquired neural features upon contact with neurogenic factors. The identity of UCB cells that acquired a neural phenotype is still unclear.


Subject(s)
Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Neurogenesis , Cell Culture Techniques , Cells, Cultured , Cytokines/metabolism , Hematopoietic Stem Cells/metabolism , Humans , Nerve Growth Factors/metabolism , Neurons/cytology , Neurons/metabolism
5.
Cytotherapy ; 20(11): 1345-1354, 2018 11.
Article in English | MEDLINE | ID: mdl-30322708

ABSTRACT

OBJECTIVE: Cell cycle plays a fundamental role in the physiology of hematopoietic stem and progenitor cells. In the present study we used a negative selection system to obtain an immature cell population-enriched for cord blood-derived CD34+ cells-and we determined its proliferation, expansion and differentiation patterns as a function of the cell cycle status. The effects of hydroxyurea (HU) were also assessed. RESULTS: As compared with cells in synthesis (S)/Gap2 (G2)/mitosis (M), cells in quiescent state (G0)/Gap1 (G1) showed a higher proliferation potential in vitro. At culture onset, G0, G1 and S/G2/M cells corresponded with 63%, 33% and 4%, respectively. Treatment with HU before culture resulted in an increase in the proportion of cells in G1 with a concomitant decrease in S/G2/M cells, without affecting the proportion of cells in G0. After 3 days of culture in the presence of recombinant cytokines, the vast majority of the cells (90%) were in G1, and by day 8, G0, G1 and S/G2/M cells corresponded with 18%, 67% and 15%, respectively. HU also induced an increase in colony-forming cell (CFC) frequency, in the proliferation and expansion capacities of cultured cells under myeloid conditions, and favored the development of the erythroid lineage. CONCLUSION: Our results show that the in vitro proliferation, expansion and differentiation potentials of immature hematopoietic cells are determined, at least in part, by their cell cycle status and that the cell cycle modifier HU significantly influences the growth of human hematopoietic cells. These results are of potential relevance for the development of ex vivo expansion protocols.


Subject(s)
Antigens, CD34/metabolism , Cell Cycle/physiology , Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Hydroxyurea/pharmacology , Blood Cells/cytology , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Colony-Forming Units Assay , Humans , Umbilical Cord/blood supply
6.
Stem Cells Transl Med ; 7(8): 602-614, 2018 08.
Article in English | MEDLINE | ID: mdl-29701016

ABSTRACT

To date, different experimental strategies have been developed for the ex vivo expansion of human hematopoietic stem (HSCs) and progenitor (HPCs) cells. This has resulted in significant advances on the use of such expanded cells in transplantation settings. To this day, however, it is still unclear to what extent those stem and progenitor cells generated in vitro retain the functional and genomic integrity of their freshly isolated counterparts. In trying to contribute to the solving of this issue, in the present study we have selected and purified three different hematopoietic cell populations: HSCs (CD34+ CD38- CD45RA- CD71- Lin- cells), myeloid progenitor cells (CD34+ CD38+ CD45RA+ CD71- Lin- cells), and erythroid progenitor cells (CD34+ CD38+ CD45RA- CD71+ Lin- cells), obtained directly from fresh human umbilical cord blood (UCB) units or generated in vitro under particular culture conditions. We, then, compared their functional integrity in vitro and their gene expression profiles. Our results indicate that in spite of being immunophenotipically similar, fresh and in vitro generated cells showed significant differences, both in functional and genetic terms. As compared to their fresh counterparts, those HSCs generated in our culture system showed a deficient content of long-term culture-initiating cells, and a marked differentiation bias toward the myeloid lineage. In addition, in vitro generated HSCs and HPCs showed a limited expansion potential. Such functional alterations correlated with differences in their gene expression profiles. These observations are relevant in terms of HSC biology and may have implications in UCB expansion and transplantation. Stem Cells Translational Medicine 2018;7:602-614.


Subject(s)
Hematopoietic Stem Cells/metabolism , Stem Cells/metabolism , Transcriptome , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, CD34/genetics , Antigens, CD34/metabolism , Cell Culture Techniques , Cells, Cultured , Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Humans , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/metabolism , Oligonucleotide Array Sequence Analysis , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , Stem Cells/cytology
7.
Int J Biochem Cell Biol ; 91(Pt A): 29-36, 2017 10.
Article in English | MEDLINE | ID: mdl-28855121

ABSTRACT

We investigated the properties of tubulin present in the sedimentable fraction ("Sed-tub") of human erythrocytes, and tracked the location and organization of tubulin in various types of cells during the process of hematopoietic/erythroid differentiation. Sed-tub was sensitive to taxol/nocodazole (drugs that modify microtubule assembly/disassembly), but was organized as part of a protein network rather than in typical microtubule form. This network had a non-uniform "connected-ring" structure, with tubulin localized in the connection areas and associated with other proteins. When tubulin was eliminated from Sed-tub fraction, this connected-ring structure disappeared. Spectrin, a major protein component in Sed-tub fraction, formed a complex with tubulin. During hematopoietic differentiation, tubulin shifts from typical microtubule structure (in pro-erythroblasts) to a disorganized structure (in later stages), and is retained in reticulocytes following enucleation. Thus, tubulin is not completely lost when erythrocytes mature; it continues to play a structural role in the Sed-tub fraction.


Subject(s)
Erythrocytes/cytology , Erythrocytes/metabolism , Hematopoiesis , Tubulin/metabolism , Adult , Blood Sedimentation/drug effects , Erythrocytes/drug effects , Female , Hematopoiesis/drug effects , Humans , Male , Nocodazole/pharmacology , Paclitaxel/pharmacology , Spectrin/metabolism , Tubulin/chemistry
8.
Stem Cells Int ; 2017: 6061729, 2017.
Article in English | MEDLINE | ID: mdl-29675046

ABSTRACT

Mesenchymal stem/stromal cells (MSCs) from bone marrow (BM) have been used in coculture systems as a feeder layer for promoting the expansion of hematopoietic progenitor cells (HPCs) for hematopoietic cell transplantation. Because BM has some drawbacks, umbilical cord blood (UCB) and placenta (PL) have been proposed as possible alternative sources of MSCs. However, MSCs from UCB and PL sources have not been compared to determine which of these cell populations has the best capacity of promoting hematopoietic expansion. In this study, MSCs from UCB and PL were cultured under the same conditions to compare their capacities to support the expansion of HPCs in vitro. MSCs were cocultured with CD34+CD38-Lin- HPCs in the presence or absence of early acting cytokines. HPC expansion was analyzed through quantification of colony-forming cells (CFCs), long-term culture-initiating cells (LTC-ICs), and CD34+CD38-Lin- cells. MSCs from UCB and PL have similar capacities to increase HPC expansion, and this capacity is similar to that presented by BM-MSCs. Here, we are the first to determine that MSCs from UCB and PL have similar capacities to promote HPC expansion; however, PL is a better alternative source because MSCs can be obtained from a higher proportion of samples.

9.
Stem Cells Dev ; 25(17): 1299-310, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27462977

ABSTRACT

The differentiation capacity, hematopoietic support, and immunomodulatory properties of human bone marrow mesenchymal stromal cells (BM-MSCs) make them attractive therapeutic agents for a wide range of diseases. Clinical scale cultures (CSCs) have been used to expand BM-MSCs for their use in cell therapy protocols; however, little is known about the functionality of the expanded cells. The main goal of the present study was to evaluate the functional characteristics of BM-MSCs expanded from CSCs to determine the quality of the cells for cellular therapy protocols. To address this issue, we analyzed the morphology, immunophenotype, differentiation potential (adipogenic, osteogenic and chondrogenic), hematopoietic support, and immunosuppressive capacity of BM-MSCs from short scale cultures (SSCs) and CSCs in a comparative manner. After 12 days of culture in CSCs (HYPERFlask System), BM-MSCs reached cell numbers of 125.52 × 10(6) ± 25.6 × 10(6) MSCs, which corresponded to the number of cells required for transplantation (∼1.7 × 10(6) MSCs/kg for a 70-kg patient). After expansion, BM-MSCs expressed the characteristic markers CD73, CD90, and CD105; however, expansion decreased their differentiation capacity toward the adipogenic, osteogenic, and chondrogenic lineages and their ability to inhibit T-cell proliferation compared with SSCs-MSCs. Importantly, CSCs-MSCs maintained the ability to support the proliferation and expansion of hematopoietic progenitor cells and the capacity to express the molecules, cytokines, and extracellular matrix proteins involved in the regulation of hematopoiesis. Our study highlights the need to evaluate the functional properties of the expanded BM-MSCs for verification of their quality for cell therapy protocols.


Subject(s)
Bone Marrow Cells/cytology , Cell Culture Techniques/methods , Cell Differentiation , Hematopoietic Stem Cells/cytology , Immunosuppression Therapy , Mesenchymal Stem Cells/cytology , Adipogenesis/genetics , Antigens, CD/metabolism , Bone Marrow Cells/metabolism , Cell Differentiation/genetics , Cell Proliferation/genetics , Cell Shape/genetics , Cells, Cultured , Chondrogenesis/genetics , Cytokines/metabolism , Extracellular Matrix/metabolism , Gene Expression Regulation , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Humans , Mesenchymal Stem Cells/metabolism
10.
Clin Dev Immunol ; 2013: 349067, 2013.
Article in English | MEDLINE | ID: mdl-24198842

ABSTRACT

Acute lymphoblastic leukemia (ALL) is the most frequent malignancy of childhood. Substantial progress on understanding the cell hierarchy within ALL bone marrow (BM) has been recorded in the last few years, suggesting that both primitive cell fractions and committed lymphoid blasts with immature stem cell-like properties contain leukemia-initiating cells. Nevertheless, the biology of the early progenitors that initiate the lymphoid program remains elusive. The aim of the present study was to investigate the ability of lymphoid progenitors from B-cell precursor ALL BM to proliferate and undergo multilineage differentiation. By phenotype analyses, in vitro proliferation assays, and controlled culture systems, the lymphoid differentiation potentials were evaluated in BM primitive populations from B-cell precursor ALL pediatric patients. When compared to their normal counterparts, functional stem and progenitor cell contents were substantially reduced in ALL BM. Moreover, neither B nor NK or dendritic lymphoid-cell populations developed recurrently from highly purified ALL-lymphoid progenitors, and their proliferation and cell cycle status revealed limited proliferative capacity. Interestingly, a number of quiescence-associated transcription factors were elevated, including the transcriptional repressor Gfi-1, which was highly expressed in primitive CD34⁺ cells. Together, our findings reveal major functional defects in the primitive hematopoietic component of ALL BM. A possible contribution of high levels of Gfi-1 expression in the regulation of the stem/progenitor cell biology is suggested.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Leukemic , Lymphoid Progenitor Cells/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Transcription Factors/genetics , Adolescent , Apoptosis , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Differentiation , Cell Proliferation , Child , Child, Preschool , Female , Humans , Infant , Lymphoid Progenitor Cells/pathology , Male , Phenotype
11.
Stem Cells Transl Med ; 2(11): 830-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24101670

ABSTRACT

Hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) play key roles in the production of mature blood cells and in the biology and clinical outcomes of hematopoietic transplants. The numbers of these cells, however, are extremely low, particularly in umbilical cord blood (UCB); thus, ex vivo expansion of human UCB-derived HSCs and HPCs has become a priority in the biomedical field. Expansion of progenitor cells can be achieved by culturing such cells in the presence of different combinations of recombinant stimulatory cytokines; in contrast, expansion of actual HSCs has proved to be more difficult because, in addition to needing recombinant cytokines, HSCs seem to deeply depend on the presence of stromal cells and/or elements that promote the activation of particular self-renewal signaling pathways. Hence, there is still controversy regarding the optimal culture conditions that should be used to achieve this. To date, UCB transplants using ex vivo-expanded cells have already been performed for the treatment of different hematological disorders, and although results are still far from being optimal, the advances are encouraging. Recent studies suggest that HSCs may also give rise to nonhematopoietic cells, such as neural, cardiac, mesenchymal, and muscle cells. Such plasticity and the possibility of producing nonhematopoietic cells at the clinical scale could bring new alternatives for the treatment of neural, metabolic, orthopedic, cardiac, and neoplastic disorders. Once standardized, ex vivo expansion of human HSCs/HPCs will surely have a positive impact in regenerative medicine.


Subject(s)
Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Stem Cells/cytology , Cytokines/metabolism , Fetal Blood/metabolism , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/metabolism , Humans , Regenerative Medicine/methods , Stem Cells/metabolism
12.
Rev. Fac. Med. UNAM ; 56(3): 47-59, may.-jun. 2013. ilus, tab
Article in Spanish | LILACS | ID: lil-725155

ABSTRACT

El conocimiento de las células troncales ha salido de las cajas petri y los animales de experimentación para llegar a áreas que habían sido inexploradas por el conocimiento biológico. Líderes religiosos, presidentes y artistas han hablado y debatido sobre ellas. Las células troncales pueden ser consideradas como celebridades biológicas y, como cualquier celebridad, son escasas y difíciles de encontrar en cualquier otro sitio que no sean los titulares periodísticos. Las células troncales poseen varias características funcionales, dentro de las que destacan su capacidad de autorenovación y su gran potencial de proliferación y de diferenciación, características que las colocan en la mira tanto de la investigación básica como la investigación traslacional o aplicada. Las células troncales se localizan en áreas muy específicas dentro de los tejidos, denominadas como nichos. Los nichos proveen a las células troncales las condiciones necesarias para regular su fisiología preservar su estado de "célula troncal", además de que participan en la regulación de su proliferación y diferenciación. El conocer la localización de las células troncales y los mecanismos que las regulan en estos sitios, nos permitirá descubrir los secretos que guardan, para conocer su papel en la fisiopatología de las enfermedades y utilizarlos como posible blanco terapéutico, sacarlas de sus nichos más eficientemente para que sean accesibles para trasplantarlas e, incluso, producir células troncales en el laboratorio e inducir su diferenciación hacia tipos celulares específicos para su uso en protocolos de terapia celular y medicina regenerativa. En esta revisión nos enfocaremos a presentar el nicho de las células troncales hematopoyéticas (CTH) y la aplicación médica de este conocimiento.


Stem cells can be considered the new celebrities in biology and medicine, reaching areas that had been unexplored by biological knowledge. Religious leaders, presidents and artists have spoken and debated about them. Like any celebrity, they are rare and hard to find anywhere else other than news headlines. Self-renewal and their vast proliferation and differentiation potentials are among some characteristics that place them in the crosshairs of both basic and translational research. Stem cells are found in very specific areas within tissues, known as niches. Stem Cell niches provide the necessary conditions to regulate their physiology, preserving their "sternness" and controlling their proliferation and differentiation. Elucidating their "zip code" will lead us to know and manipulate their regulatory mechanisms. The stem cell niche is emerging as a new therapeutic target. We will discuss the hematopoietic stem cell niche and the clinical application of this knowledge.

13.
Transfusion ; 53(3): 668-78, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22845003

ABSTRACT

BACKGROUND: Ex vivo expansion of hematopoietic stem and progenitor cells has become a priority in the experimental hematology arena. In this study we have obtained different hematopoietic cell populations from umbilical cord blood and simultaneously assessed their proliferation and expansion kinetics. Our main goal was to determine which one of these cell populations would be more suitable for clinical-grade ex vivo expansion. STUDY DESIGN AND METHODS: By using immunomagnetic-negative selection and cell sorting, five cell populations were obtained: unseparated mononuclear cells (MNCs; I); two lineage-negative cell populations, one enriched for CD34+ CD38+ cells (II) and the other enriched for CD34+ CD38- cells (III); and two CD34+ cell fractions purified by fluorescence-activated cell sorting, one containing CD34+ CD38+ cells (IV) and the other containing CD34+ CD38- cells (V). The kinetics of such populations were analyzed in both relative and absolute terms. RESULTS: No expansion was observed in Population I; in contrast, significant increments in the numbers of both progenitor and stem cells were observed in cultures of Populations II to V. Population V (reaching 12,800-fold increase in total cells; 1280-fold increase in CD34+ cells; 490-fold increase in colony-forming cells; and 12-fold increase in long-term culture-initiating cells) showed the highest proliferation and expansion potentials. CONCLUSION: Our study suggests that the cell fraction containing greater than 98% CD34+ CD38- cells would be the ideal one for large-scale ex vivo expansion; however, based on our data, it seems that, except for MNCs, all other cell populations could also be used as input cell fractions.


Subject(s)
Cell Culture Techniques , Cell Proliferation , Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Antigens, CD34/metabolism , Cell Count , Cell Culture Techniques/methods , Cell Separation , Cells, Cultured , Choice Behavior , Colony-Forming Units Assay/methods , Fetal Blood/physiology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/physiology , Humans , Primary Cell Culture/methods , Time Factors
14.
Blood Cells Mol Dis ; 47(4): 205-13, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-21911304

ABSTRACT

In trying to contribute to our knowledge on the role of Notch and its ligands within the human hematopoietic system, we have assessed the effects of the OP9 stroma cell line - naturally expressing Jagged-1 - transduced with either the Delta-1 gene (OP9-DL1 cells) or with vector alone (OP9-V), on the in vitro growth of two different hematopoietic cell populations. Primitive (CD34(+) CD38(-) Lin(-)) and intermediate (CD34(+) CD38(+) Lin(-)) CD34(+) cell subsets from human cord blood were cultured in the presence of 7 stimulatory cytokines under four different conditions: cytokines alone (control); cytokines and mesenchymal stromal cells; cytokines and OP9-V cells; cytokines and OP9-DL1 cells. Proliferation and expansion were determined after 7days of culture. Culture of CD34(+) CD38(-) Lin(-) cells in the presence of OP9-V or OP9-DL1 cells resulted in a significant increase in the production of new CD34(+) CD38(-) Lin(-) cells (expansion), which expressed increased levels of Notch-1; in contrast, production of total nucleated cells (proliferation) was reduced, as compared to control conditions. In cultures of CD34(+) CD38(+) Lin(-) cells established in the presence of OP9-V or OP9-DL1 cells, expansion was similar to that observed in control conditions, whereas proliferation was also reduced. Interestingly, in these latter cultures we observed production of CD34(+) CD38(-) Lin(-) cells. Our results indicate that, as compared to MSC, OP9 cells were more efficient at inducing self-renewal and/or de novo generation of primitive (CD34(+) CD38(-) Lin(-)) cells, and suggest that such effects were due, at least in part, to the presence of Jagged-1 and DL1.


Subject(s)
Antigens, CD34/analysis , Calcium-Binding Proteins/metabolism , Fetal Blood/cytology , Hematopoietic Stem Cells/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , ADP-ribosyl Cyclase 1/analysis , Adolescent , Adult , Cell Line , Cell Proliferation , Cells, Cultured , Hematopoietic Stem Cells/cytology , Humans , Jagged-1 Protein , Ligands , Receptors, Notch/metabolism , Serrate-Jagged Proteins , Stromal Cells/metabolism , Young Adult
15.
Leuk Res ; 35(2): 250-5, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20621354

ABSTRACT

In the present study we have determined the content of hematopoietic and stromal progenitors in multiple myeloma (MM) bone marrow, and assessed their in vitro growth. Marrow cells were obtained from 17 MM patients at the time of diagnosis, and from 6 hematologically normal subjects. When mononuclear cells (MNC) from MM marrow were cultured, reduced numbers of hematopoietic progenitors were detected and their growth in long-term cultures was deficient, as compared to cultures of normal cells. When cell fractions enriched for CD34(+) Lin(-) cells were obtained, the levels of hematopoietic progenitors from MM marrow were within the normal range, and so was their growth kinetics in liquid suspension cultures. The levels of fibroblast progenitors in MM were not statistically different from those in normal marrow; however, their proliferation potential was significantly reduced. Conditioned media from MM-derived MNC and stroma cells contained factors that inhibited normal progenitor cell growth. Our observations suggest that hematopoietic progenitors in MM marrow are intrinsically normal; however, their growth in LTMC may be hampered by the presence of abnormal accessory and stroma cells. These results suggest that besides its role in the generation of osteolytic lesions and the expansion of the myeloma clone, the marrow microenvironment in MM may have a negative effect on hematopoiesis.


Subject(s)
Bone Marrow Cells/pathology , Hematopoietic Stem Cells/pathology , Multiple Myeloma/pathology , Aged , Cell Proliferation , Female , Fibroblasts/pathology , Humans , In Vitro Techniques , Male , Middle Aged , Stromal Cells/pathology
16.
Cytotherapy ; 11(7): 886-96, 2009.
Article in English | MEDLINE | ID: mdl-19903101

ABSTRACT

BACKGROUND AIMS: We have previously characterized the in vitro growth of two cord blood-derived hematopoietic cell populations in liquid cultures supplemented with recombinant cytokines. In the present study, we assessed the effects of bone marrow-derived mesenchymal stromal cells (MSC) on the growth of such cells. METHODS: CD34(+) CD38(+) Lin(-) and CD34(+) CD38(-) Lin(-) cells were obtained by negative selection, and cultured in the presence of marrow-derived MSC and/or early- and late-acting cytokines. Hematopoietic cell growth was assessed throughout a 30-day culture period. RESULTS: In the presence of MSC alone, both populations showed significant proliferation. Direct contact between MSC and CD34(+) cells was fundamental for optimal growth, especially for CD34(+) CD38(-) Lin(-) cells. In the presence of early-acting cytokines alone, cell growth was significantly higher than in cultures established with MSC but no cytokines. In cultures containing both MSC and early-acting cytokines, a further stimulation was observed only for CD34(+) CD38(-) Lin(-) cells. The cytokine cocktail containing both early- and late-acting cytokines was significantly more potent at inducing hematopoietic cell growth than the early-acting cytokine cocktail. When cultures were supplemented with early- and late-acting cytokines, MSC had no further effect on the growth of hematopoietic cells. CONCLUSIONS: MSC seem to play a key role, particularly on more primitive (CD34(+) CD38(-) Lin(-)) cells, only in the absence of cytokines or the presence of early-acting cytokines. When both early- and late-acting cytokines are present in culture, MSC seem to be unnecessary for optimal development of CFC and CD34(+) cells.


Subject(s)
Bone Marrow Cells/metabolism , Cytokines/pharmacology , Hematopoietic Stem Cells/metabolism , Mesenchymal Stem Cells/metabolism , Recombinant Proteins/pharmacology , ADP-ribosyl Cyclase 1/biosynthesis , Antigens, CD34/biosynthesis , Bone Marrow Cells/cytology , Cell Growth Processes , Coculture Techniques , Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Humans , Immunization , Mesenchymal Stem Cells/cytology , Stromal Cells/cytology
17.
Leuk Res ; 32(9): 1407-16, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18405968

ABSTRACT

Two different reports, including one from our own group, have recently demonstrated the presence of severe chromosomal abnormalities in mesenchymal stem cells (MSC) from patients with myelodysplastic syndromes (MDS). In the present study, we have assessed whether such cytogenetic abnormalities result in functional deficiencies in vitro. We found that both normal and MDS MSC showed similar expression patterns of cell adhesion molecules and extracellular matrix proteins. MDS MSC layers showed the capability to differentiate towards adipocytes, chondrocytes and osteoblasts, and supported the growth of early umbilical cord blood progenitors in a co-culture system. Unstimulated MDS MSC secreted more IL-1beta and after treatment with TNFalpha, they secreted more SCF, as compared to their normal counterparts. The present study demonstrates that, in spite of harboring severe chromosomal alterations, most of the functional properties of MDS-derived MSC remain normal, including their ability to support normal hematopoiesis in vitro.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cells/metabolism , Myelodysplastic Syndromes/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Adolescent , Adult , Cell Adhesion Molecules/metabolism , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Coculture Techniques , Colony-Forming Units Assay , Cytogenetic Analysis , Extracellular Matrix Proteins/metabolism , Fetal Blood/cytology , Fetal Blood/metabolism , Humans , Interleukin-1beta/metabolism , Middle Aged , Myelodysplastic Syndromes/pathology , Osteoblasts/cytology , Osteoblasts/metabolism , Tumor Necrosis Factor-alpha/metabolism
18.
Leuk Lymphoma ; 47(7): 1379-86, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16923572

ABSTRACT

One has previously characterized two different hematopoietic cell populations (obtained by negative-selection) from normal bone marrow. Population I was enriched for CD34+ Lin- cells, whereas Population II was enriched for CD34+ CD38- Lin- cells. Both populations showed elevated proliferation and expansion potentials in serum-free liquid cultures, supplemented with a combination of eight different cytokines, with the latter displaying more immature features than the former. One has also characterized the chronic myeloid leukemia (CML) counterparts of these two populations and demonstrated functional deficiencies in terms of their growth in culture. In keeping with this line of research, the goal of the present study was to obtain the same two populations (Populations I and II) from acute myeloid leukemia (AML) bone marrow and to characterize their biological behavior under the same culture conditions. The results demonstrated that AML-derived Populations I and II were unable to proliferate in culture conditions that allowed significant proliferation of Populations I and II from normal marrow. Population I from AML also showed a deficient expansion capacity; in contrast, Population II cells were able to expand to a similar extent to the one observed for Population II from normal marrow. Both normal and AML populations were highly sensitive to the inhibitory effects of TNF-alpha; interestingly, whereas in normal fractions TNF-alpha showed a more pronounced inhibitory effect on more mature cells (Population I), this cytokine inhibited proliferation and expansion of AML Populations I and II in a similar degree. It is noteworthy that the functional deficiencies observed in AML cells were even more pronounced than those previously reported for cultures of CML cells. The results reported here may be of relevance considering the interest by several groups in developing methods for the in vitro purging of leukemic cells, as part of protocols for autologous transplantation of hematopoietic cells in leukemic patients.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Marrow/metabolism , Cytokines/metabolism , Leukemia, Myeloid, Acute/metabolism , ADP-ribosyl Cyclase 1/biosynthesis , Adolescent , Adult , Antigens, CD34/biosynthesis , Cell Culture Techniques/methods , Cell Proliferation , Culture Media, Serum-Free , Female , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Male , Middle Aged , Stem Cells/metabolism
19.
Leuk Lymphoma ; 45(6): 1247-54, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15360008

ABSTRACT

We have previously demonstrated that when cultured in Dexter-type Long-Term Marrow Cultures (LTMC), hematopoietic progenitor cells (HPC) from patients with Diffuse Large B-Cell Lymphoma (DLBCL) show a defective proliferation, as compared to HPC from normal marrow. In that study it was also demonstrated that functional alterations were present in the hematopoietic microenvironment developed in culture; thus, it was not clear whether such a defective proliferation in vitro was due to an intrinsic defect in the HPC compartment of DLBCL patients, or to an altered microenvironment, or both. In order to address this question, in the present study we have assessed the proliferation and expansion potentials of HPC present in bone marrow from patients with DLBCL, in cytokine-supplemented liquid cultures initiated with a cell population enriched for CD34+ Lin- cells, in the absence of stromal cells and in the presence of reduced numbers of accessory cells. Our results demonstrated that bone marrow-derived HPC from patients with DLBCL, both before and right after chemotherapy, possessed reduced proliferation and expansion potentials in vitro, as compared to their normal counterparts. Interestingly, in patients analyzed 18 months after treatment the proliferation and expansion levels were similar to those of normal HPC, indicating a complete restoration of the hematopoietic function. Although the reason for these observations is not clear, our results suggest the possibility that primitive CD34+ progenitor cells present in bone marrow, which show deficient proliferation and expansion potentials in vitro, are involved in the origin/progression of DLBCL.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Marrow/pathology , Hematopoietic Stem Cells/pathology , Lymphoma, B-Cell/drug therapy , Lymphoma, Large B-Cell, Diffuse/drug therapy , Adult , Aged , Antigens, CD34/metabolism , Cells, Cultured , Colony-Forming Units Assay , Female , Humans , In Vitro Techniques , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Male , Middle Aged , Stromal Cells/metabolism
20.
Stem Cells Dev ; 13(4): 382-9, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15345132

ABSTRACT

In the present study, we have assessed, in a comparative manner, the in vitro proliferation and expansion potentials of hematopoietic progenitor cells (HPC) present in mobilized peripheral blood from normal subjects (MPB-n; n = 18) and cancer patients (MPB-c; n = 18). The latter included patients with breast cancer (BrCa; n = 8), Hodgkin disease (HD; n = 4), non-Hodgkin lymphoma (NHL; n = 3), and acute myeloid leukemia (AML; n = 3). Progenitor cells from normal bone marrow (BM) and umbilical cord blood (UCB) were included as controls. HPC, enriched by a negative selection procedure, were cultured for 25 days, in serum-free liquid media in the presence of a cytokine combination including early- and late-acting cytokines. Our results demonstrate that the in vitro biological properties of progenitor cells present in MPB differ, depending on whether they are derived from healthy individuals, from patients with solid tumors, or from patients with hematological neoplasias. Among all cell sources analyzed, UCB-derived progenitors showed the greatest proliferation and expansion potentials (1000-fold increase in total cell numbers on day 15, and a 22-fold increase in myeloid progenitor cell numbers, at day 10). Progenitor cells present in MPB from hematologically normal individuals showed proliferation and expansion potentials comparable to those of HPC from normal BM (500-fold increase in total cell numbers on day 15, and a 14-fold increase in myeloid progenitor cell numbers, at day 10). The proliferation/expansion potentials of MPB progenitors from BrCa patients were also within the normal range, although in the lower levels (327-fold increase in total cell numbers, on day 15, and 11.8-fold increase in myeloid progenitors, at day 10). In contrast, progenitors present in MPB from patients with HD, NHL, and especially AML, showed reduced in vitro capacities (119-, 102-, and 51-fold increase in total cell numbers, respectively; and 8-, 4-, and 2.6-fold increase in myeloid progenitor cells, respectively). To our knowledge, this is the first report in which the in vitro proliferation and expansion potentials of HPC from MPB from normal subjects and cancer patients are assessed simultaneously in a comparative manner.


Subject(s)
Bone Marrow Cells/cytology , Cell Division/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/pathology , Neoplasms/pathology , Neoplasms/therapy , Adolescent , Adult , Antigens, CD/blood , Antigens, CD34/blood , Cell Culture Techniques/methods , Humans , Kinetics , Middle Aged , Reference Values , Stem Cell Transplantation , Transplantation, Autologous
SELECTION OF CITATIONS
SEARCH DETAIL
...