Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Blood ; 127(26): 3398-409, 2016 06 30.
Article in English | MEDLINE | ID: mdl-27114459

ABSTRACT

Recently, interactions between thrombopoietin (TPO) and its receptor, the myeloproliferative leukemia (MPL) virus oncogene, have been shown to play a role in the development and progression of myeloproliferative neoplasms including myelofibrosis (MF). These observations have led to the development of strategies to disrupt the association of TPO with its receptor as a means of targeting MF hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs). In this report, we show that although both splenic and peripheral blood MF CD34(+) cells expressed lower levels of MPL than normal CD34(+) cells, TPO promoted the proliferation of MF CD34(+) cells and HPCs in a dose-dependent fashion. Furthermore, the treatment of MF but not normal CD34(+) cells with a synthesized MPL antagonist, LCP4, decreased the number of CD34(+)Lin(-) cells and all classes of assayable HPCs (colony-forming unit-megakaryocyte [CFU-MK], CFU-granulocyte/macrophage, burst-forming unit-erythroid/CFU-erythroid, and CFU-granulocyte/erythroid/macrophage/MK) irrespective of their mutational status. In addition, LCP4 treatment resulted in the depletion of the number of MF HPCs that were JAK2V617F(+) Moreover, the degree of human cell chimerism and the proportion of malignant donor cells were significantly reduced in immunodeficient mice transplanted with MF CD34(+) cell grafts treated with LCP4. These effects of LCP4 on MF HSCs/HPCs were associated with inhibition of JAK-STAT activity, leading to the induction of apoptosis. These findings demonstrate that such specific anti-cytokine receptor antagonists represent a new class of drugs that are capable of targeting MF HSCs.


Subject(s)
Hematopoietic Stem Cells/metabolism , Primary Myelofibrosis/drug therapy , Receptors, Thrombopoietin/antagonists & inhibitors , Aged , Amino Acid Substitution , Animals , Antigens, CD34/genetics , Antigens, CD34/metabolism , Female , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/pathology , Heterografts , Humans , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Male , Mice , Middle Aged , Mutation, Missense , Primary Myelofibrosis/genetics , Primary Myelofibrosis/metabolism , Primary Myelofibrosis/pathology , Receptors, Thrombopoietin/genetics , Receptors, Thrombopoietin/metabolism
2.
Biopolymers ; 2018 Jan 30.
Article in English | MEDLINE | ID: mdl-29381187

ABSTRACT

A 54-member library of boronated octapeptides, with all but the boronated residue being proteinogenic, was tested for affinity to a set of saccharides commonly found on the terminus of mammalian glycans. After experimentation with a high-throughput dye-displacement assay, attention was focused on isothermal titration calorimetry as a tool to provide reliable affinity data, including enthalpy and entropy of binding. A small number of boronated peptides showed higher affinity and significant selectivity for N-acetylneuraminic acid over methyl-α-d-galactopyranoside, methyl-α/ß-l-fucopyranoside and N-acetyl-d-glucosamine. Thermodynamic data showed that for most of the boronated peptides studied, saccharide binding was associated with a significant increase in entropy, presumably resulting from the displacement of semiordered water molecules from around the sugar and/or peptide.

3.
Platelets ; 28(7): 649-656, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28067095

ABSTRACT

Blood transfusion services face an ever-increasing demand for donor platelets to meet clinical needs. Whilst strategies for increasing platelet storage life and improving the efficiency of donor platelet collection are important, in the longer term, platelets generated by bio-manufacturing processes will be required to meet demands. Production of sufficient numbers of in vitro-derived platelets for transfusion represents a significant bioengineering challenge. In this review, we highlight recent progress in this area of research and outline the main technical and biological obstacles that need to be met before this becomes feasible and economic. A critical consideration is assurance of the functional properties of these cells as compared to their fresh, donor collected, counterparts. We contend that platelet-like particles and in vitro-derived platelets that phenotypically resemble fresh platelets must deliver the same functions as these cells upon transfusion. We also note recent progress with immortalized megakaryocyte progenitor cell lines, molecular strategies for reducing expression of HLA Class I to generate universal donor platelets and the move to early clinical studies with in vitro-derived platelets.


Subject(s)
Blood Platelets/cytology , Cell Culture Techniques , Megakaryocytes/cytology , Platelet Transfusion/standards , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/immunology , Blood Platelets/immunology , Cell Dedifferentiation/drug effects , Cell Differentiation/drug effects , Cell Line, Transformed , Cytokines/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/immunology , Gene Silencing , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/immunology , Intercellular Signaling Peptides and Proteins/pharmacology , Megakaryocytes/drug effects , Megakaryocytes/immunology , Microfluidics/instrumentation , Microfluidics/methods , Platelet Transfusion/statistics & numerical data
4.
Stem Cells ; 33(7): 2351-7, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25865259

ABSTRACT

Factor V (FV) and factor X (FX) activate and complex to form prothrombinase which subsequently cleaves prothrombin (PT), converting it to active thrombin. Thrombin cleaved osteopontin (tcOPN) contains a cryptic binding site for α4 ß1 and α9 ß1 integrins. We have previously shown that hematopoietic stem cells (HSC) bind to tcOPN via this site resulting in a decrease in their proliferation and differentiation. Therefore, tcOPN and the factors required for its generation are important components of the HSC niche. Herein we show mature megakaryocytes (MM, ≥8N) contain FV, FX, and PT mRNA and protein. Furthermore, we show 8N, 16N, 32N, and 64N MM all release the required factors to enable thrombin cleavage of OPN. Importantly, mice devoid of the myeloproliferative leukemia protein (Mpl), c-Mpl(-/-) mice, contain only approximately 10% of normal megakaryocyte numbers, showed significantly reduced FX and tcOPN protein levels in endosteal bone marrow (BM). In addition, WT hematopoietic progenitors and HSC showed reduced homing to the BM of c-Mpl(-/-) mice. This is the first report identifying MM as a key cellular component in the production of tcOPN in situ, allowing the BM microenvironment to self regulate HSC biology via tcOPN.


Subject(s)
Bone Marrow/metabolism , Hematopoietic Stem Cells/metabolism , Megakaryocytes/metabolism , Osteopontin/metabolism , Thrombin/metabolism , Animals , Cell Differentiation , Cell Movement , Megakaryocytes/cytology , Mice , Stem Cell Niche , Tumor Microenvironment
5.
Org Biomol Chem ; 12(6): 965-78, 2014 Feb 14.
Article in English | MEDLINE | ID: mdl-24363056

ABSTRACT

The α9ß1 and α4ß1 integrin subtypes are expressed on bone marrow haemopoietic stem cells and have important roles in stem cell regulation and trafficking. Although the roles of α4ß1 integrin have been thoroughly investigated with respect to HSC function, the role of α9ß1 integrin remains poorly characterised. Small molecule fluorescent probes are useful tools for monitoring biological processes in vivo, to determine cell-associated protein localisation and activation, and to elucidate the mechanism of small molecule mediated protein interactions. Herein, we report the design, synthesis and integrin-dependent cell binding properties of a new fluorescent α9ß1 integrin antagonist (R-BC154), which was based on a series of N-phenylsulfonyl proline dipeptides and assembled using the Cu(I)-catalyzed azide alkyne cycloaddition (CuAAC) reaction. Using transfected human glioblastoma LN18 cells, we show that R-BC154 exhibits high nanomolar binding affinities to α9ß1 integrin with potent cross-reactivity against α4ß1 integrin under physiological mimicking conditions. On-rate and off-rate measurements revealed distinct differences in the binding kinetics between α9ß1 and α4ß1 integrins, which showed faster binding to α4ß1 integrin relative to α9ß1, but more prolonged binding to the latter. Finally, we show that R-BC154 was capable of binding rare populations of bone marrow haemopoietic stem and progenitor cells when administered to mice. Thus, R-BC154 represents a useful multi-purpose fluorescent integrin probe that can be used for (1) screening small molecule inhibitors of α9ß1 and α4ß1 integrins; (2) investigating the biochemical properties of α9ß1 and α4ß1 integrin binding and (3) investigating integrin expression and activation on defined cell phenotypes in vivo.


Subject(s)
Bone Marrow Cells/cytology , Dipeptides/pharmacology , Drug Design , Fluorescent Dyes/pharmacology , Integrin alpha4beta1/antagonists & inhibitors , Integrins/antagonists & inhibitors , Proline/pharmacology , Rhodamines/pharmacology , Binding Sites/drug effects , Cell Line, Tumor , Dipeptides/chemical synthesis , Dipeptides/chemistry , Dose-Response Relationship, Drug , Fluorescence , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , Humans , Molecular Conformation , Proline/analogs & derivatives , Proline/chemistry , Rhodamines/chemical synthesis , Rhodamines/chemistry , Structure-Activity Relationship
6.
Blood ; 118(6): 1516-24, 2011 Aug 11.
Article in English | MEDLINE | ID: mdl-21673348

ABSTRACT

A large body of evidence suggests hemopoietic stem cells (HSCs) exist in an endosteal niche close to bone, whereas others suggest that the HSC niche is intimately associated with vasculature. In this study, we show that transplanted hemopoietic stem and progenitor cells (HSPCs) home preferentially to the trabecular-rich metaphysis of the femurs in nonablated mice at all time points from 15 minutes to 15 hours after transplantation. Within this region, they exist in an endosteal niche in close association with blood vessels. The preferential homing of HSPCs to the metaphysis occurs rapidly after transplantation, suggesting that blood vessels within this region may express a unique repertoire of endothelial adhesive molecules. One candidate is hyaluronan (HA), which is highly expressed on the blood vessel endothelium in the metaphysis. Analysis of the early stages of homing and the spatial dis-tribution of transplanted HSPCs at the single-cell level in mice devoid of Has3-synthesized HA, provides evidence for a previously undescribed role for HA expressed on endothelial cells in directing the homing of HSPCs to the metaphysis.


Subject(s)
Blood Vessels/cytology , Bone Marrow/blood supply , Bone and Bones/cytology , Hematopoietic Stem Cells/cytology , Animals , Blood Vessels/metabolism , Blood Vessels/ultrastructure , Bone and Bones/metabolism , Cell Adhesion Molecules/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Female , Femur/cytology , Femur/metabolism , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Hyaluronan Synthases , Hyaluronic Acid/metabolism , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Stem Cell Niche/blood supply , Stem Cell Niche/cytology , Transendothelial and Transepithelial Migration , X-Ray Microtomography
7.
Blood ; 116(17): 3185-96, 2010 Oct 28.
Article in English | MEDLINE | ID: mdl-20631378

ABSTRACT

Hemopoietic stem cells (HSCs) reside within a specified area of the bone marrow (BM) cavity called a "niche" that modulates HSC quiescence, proliferation, differentiation, and migration. Our previous studies have identified the endosteal BM region as the site for the HSC niche and demonstrated that hemopoietic stem and progenitor populations (HSPCs, LSK) isolated from different BM regions exhibit significantly different hemopoietic potential. In this study, we have analyzed subpopulations of LSK cells isolated from different regions of the BM and showed that CD150(+)CD48(-)LSK HSCs within the endosteal BM region have superior proliferative capacity and homing efficiency compared with CD150(+)CD48(-)LSK HSCs isolated from the central BM. Furthermore, we show, for the first time, that a subset of CD150(+)CD48(+)LSK progenitor cells, previously defined as B-lymphoid primed hemopoietic cells, are capable of multilineage reconstitution, however, only when isolated from the endosteal region. In addition, we provide evidence for an unrecognized role of CD48 in HSC homing. Together, our data provide strong evidence that highly purified HSCs show functional differences depending on their origin within the BM and that the most primitive HSCs reside within the endosteal BM region.


Subject(s)
Antigens, CD/metabolism , Bone Marrow/anatomy & histology , Cell Proliferation , Hematopoietic Stem Cells/cytology , Proto-Oncogene Proteins c-kit/metabolism , Receptors, Cell Surface/metabolism , Animals , Antigens, Ly/metabolism , CD48 Antigen , Cell Cycle , Hematopoietic Stem Cell Transplantation , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Signaling Lymphocytic Activation Molecule Family Member 1
8.
Cytokine ; 58(2): 218-25, 2012 May.
Article in English | MEDLINE | ID: mdl-22341722

ABSTRACT

Granulocyte colony stimulating factor (G-CSF) is clinically well established for the mobilization of hematopoietic stem cells (HSC). Extensive data on the underlying mechanism of G-CSF induced mobilization is available; however, little is known regarding the functional effect of G-CSF on HSC within the bone marrow (BM). In this study we analyzed the proportion and number of murine HSC in the endosteal and central bone marrow regions after 4 days of G-CSF administration. We demonstrate that the number of HSC, defined as CD150(+)CD48(-)LSK cells (LSKSLAM cells), increased within the central BM region in response to G-CSF, but not within the endosteal BM region. In addition the level of CD150 and CD48 expression also increased on cells isolated from both regions. We further showed that G-CSF mobilized proportionally fewer LSKSLAM compared to LSK cells, mobilized LSKSLAM had colony forming potential and the presence of these cells can be used as a measure for mobilization efficiency. Together we provide evidence that HSC in the BM respond differently to G-CSF and this is dependent on their location. These findings will be valuable in developing new agents which specifically mobilize HSC from the endosteal BM region, which we have previously demonstrated to have significantly greater hematopoietic potential compared to their phenotypically identical counterparts located in other regions of the BM.


Subject(s)
Bone Marrow/drug effects , Cell Division/drug effects , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/cytology , Animals , Antigens, CD/immunology , CD48 Antigen , Cell Cycle , Flow Cytometry , Hematopoietic Stem Cells/immunology , Mice , Mice, Inbred C57BL , Receptor-Like Protein Tyrosine Phosphatases, Class 3/immunology
9.
Blood ; 114(1): 49-59, 2009 Jul 02.
Article in English | MEDLINE | ID: mdl-19417209

ABSTRACT

Osteopontin (OPN), a multifunctional acidic glycoprotein, expressed by osteoblasts within the endosteal region of the bone marrow (BM) suppresses the proliferation of hemopoietic stem and progenitor cells and also regulates their lodgment within the BM after transplantation. Herein we demonstrate that OPN cleavage fragments are the most abundant forms of this protein within the BM. Studies aimed to determine how hemopoietic stem cells (HSCs) interact with OPN revealed for the first time that murine and human HSCs express alpha(9)beta(1) integrin. The N-terminal thrombin cleavage fragment of OPN through its binding to the alpha(9)beta(1) and alpha(4)beta(1) integrins plays a key role in the attraction, retention, regulation, and release of hemopoietic stem and progenitor cells to, in, and from their BM niche. Thrombin-cleaved OPN (trOPN) acts as a chemoattractant for stem and progenitor cells, mediating their migration in a manner that involves interaction with alpha(9)beta(1) and alpha(4)beta(1) integrins. In addition, in the absence of OPN, there is an increased number of white blood cells and, specifically, stem and progenitor cells in the peripheral circulation.


Subject(s)
Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Integrin alpha4beta1/metabolism , Integrins/metabolism , Osteopontin/physiology , Animals , Base Sequence , CHO Cells , Cell Line , Chemotaxis/drug effects , Chemotaxis/physiology , Cricetinae , Cricetulus , DNA Primers/genetics , Fetal Blood/cytology , Gene Expression , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoiesis/drug effects , Hematopoiesis/genetics , Hematopoiesis/physiology , Hematopoietic Stem Cells/drug effects , Humans , In Vitro Techniques , Integrin alpha4beta1/genetics , Integrins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Osteopontin/deficiency , Osteopontin/genetics , Osteopontin/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thrombin/metabolism
10.
Blood ; 114(23): 4859-70, 2009 Nov 26.
Article in English | MEDLINE | ID: mdl-19805619

ABSTRACT

Deregulated cell survival programs are a classic hallmark of cancer. We have previously identified a serine residue (Ser585) in the betac subunit of the granulocyte-macrophage colony-stimulating factor receptor that selectively and independently promotes cell survival. We now show that Ser585 phosphorylation is constitutive in 20 (87%) of 23 acute myeloid leukemia (AML) patient samples, indicating that this survival-only pathway is frequently deregulated in leukemia. We performed a global expression screen to identify gene targets of this survival pathway and report a 138-gene betac Ser585-regulated transcriptome. Pathway analysis defines a gene network enriched for PI3-kinase target genes and a cluster of genes involved in cancer and cell survival. We show that one such gene, osteopontin (OPN), is a functionally relevant target of the Ser585-survival pathway as shown by siRNA-mediated knockdown of OPN expression that induces cell death in both AML blasts and CD34(+)CD38(-)CD123(+) leukemic progenitors. Increased expression of OPN at diagnosis is associated with poor prognosis with multivariate analysis indicating that it is an independent predictor of overall patient survival in normal karyotype AML (n = 60; HR = 2.2; P = .01). These results delineate a novel cytokine-regulated Ser585/PI3-kinase signaling network that is deregulated in AML and identify OPN as a potential prognostic and therapeutic target.


Subject(s)
Gene Expression Profiling , Hematopoietic Stem Cells/metabolism , Leukemia, Myeloid/genetics , Neoplasm Proteins/physiology , Osteopontin/physiology , Adult , Aged , Cell Survival , Cytokine Receptor Common beta Subunit/metabolism , Female , Gene Expression Regulation, Leukemic , Gene Knockdown Techniques , Gene Regulatory Networks , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/pathology , Humans , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/mortality , Leukemia, Myeloid/pathology , Male , Middle Aged , Neoplasm Proteins/genetics , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Osteopontin/biosynthesis , Osteopontin/genetics , Phosphatidylinositol 3-Kinases/physiology , Phosphoinositide-3 Kinase Inhibitors , Phosphoserine/metabolism , Prognosis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , RNA, Small Interfering/pharmacology , Signal Transduction/genetics , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/pathology
11.
Trials ; 21(1): 474, 2020 Jun 04.
Article in English | MEDLINE | ID: mdl-32493459

ABSTRACT

OBJECTIVES: Objective: To undertake a pilot, feasibility RCT of umbilical cord blood derived cell therapy for treatment of adult patients infected with SARS-CoV-2 virus related moderate-to-severe pneumonia to prevent progression to severe ARDS. HYPOTHESIS: Expanded cord blood derived cell therapy will be feasible, well tolerated and show potential efficacy in the treatment of acute COVID-19 related moderate to severe pneumonia in adult patients because of their powerful anti-inflammatory and immunomodulatory properties. TRIAL DESIGN: Pilot, parallel design randomised controlled trial. PARTICIPANTS: The trial will recruit 24 hospitalised patients with confirmed SARS-CoV-2 infection and pneumonia from July to December 2020 at Monash Medical Centre in Melbourne, Australia. INTERVENTION AND COMPARATOR: Intervention: Intravenous injection of expanded umbilical cord blood cells at a dose of 5 million cells/kg (maximum dose - 500 million cells). Cell infusion will occur over 30-60 minutes through a peripheral intravenous cannula. Standard supportive care will continue as needed. Comparator: Standard supportive care. MAIN OUTCOMES: Safety and tolerability of cell administration within first 24 hours of administration; clinical improvement on a seven-category clinical improvement ordinal scale. RANDOMISATION: Randomisation will be done using computer generated allocation to intervention/ control groups in a 1:1 ratio (in blocks of 6) using sealed opaque envelopes. BLINDING (MASKING): This will be an unblinded study, given that it is the first study using expanded cord blood cells in COVID-19 patients. There will be no placebo infusion. NUMBERS TO BE RANDOMISED (SAMPLE SIZE): Twelve participants in each group. Total n=24. TRIAL STATUS: CBC-19 protocol v2, dated 23rd April 2020. Recruitment has not started yet. Estimated recruitment timeline is between 1st July - 31st December 2020. TRIAL REGISTRATION: Australian New Zealand Clinical Trials Registry, ACTRN12620000478910, registered 16th April 2020. FULL PROTOCOL: The full protocol is attached as an additional file, accessible from the Trials website (Additional file 1). In the interest in expediting dissemination of this material, the familiar formatting has been eliminated; this Letter serves as a summary of the key elements of the full protocol.


Subject(s)
Betacoronavirus/pathogenicity , Cord Blood Stem Cell Transplantation , Coronavirus Infections/surgery , Pneumonia, Viral/surgery , COVID-19 , Cord Blood Stem Cell Transplantation/adverse effects , Coronavirus Infections/diagnosis , Coronavirus Infections/virology , Disease Progression , Host-Pathogen Interactions , Humans , Pandemics , Pilot Projects , Pneumonia, Viral/diagnosis , Pneumonia, Viral/virology , Randomized Controlled Trials as Topic , SARS-CoV-2 , Time Factors , Treatment Outcome , Victoria
12.
Biol Blood Marrow Transplant ; 14(6): 603-21, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18489986

ABSTRACT

The recent explosion in the understanding of the cellular and molecular mechanisms underlying hematopoietic stem and progenitor cell (HSPC) mobilization has facilitated development of novel therapeutic agents, targeted at improving mobilization kinetics as well as HSPC yield. With the development of new agents comes the challenge of choosing efficient and relevant preclinical studies for the testing of the HSPC mobilization efficacy of these agents. This article reviews the use of the mouse as a convenient small animal model of HSPC mobilization and transplantation, and outlines the range of murine assays that can be applied to assess novel HSPC mobilizing agents. Techniques to demonstrate murine HSPC mobilization are discussed, as well as the role of murine assays to confirm human HSPC mobilization, and techniques to investigate the biologic phenotype of HSPC mobilized by these novel agents. Technical aspects regarding mobilization regimens and control arms, and choice of experimental animals are also discussed.


Subject(s)
Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Models, Animal , Animals , Animals, Congenic , Cell Culture Techniques/methods , Cell Lineage , Cells, Cultured/drug effects , Drug Evaluation, Preclinical/methods , Female , Graft Survival , Hematopoietic Cell Growth Factors/pharmacology , Hematopoietic Stem Cells/classification , Hematopoietic Stem Cells/cytology , Humans , Male , Mice , Mice, Inbred NOD , Mice, Inbred Strains , Mice, SCID , Myeloablative Agonists/pharmacology , Radiation Chimera
13.
Eur J Haematol ; 80(1): 20-30, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18028434

ABSTRACT

OBJECTIVE: Hematopoietic progenitor cells (HPC) as well as tissue committed stem cells expressing mRNA specific to various somatic tissues are thought to be part of the CD34+ bone marrow compartment. In this study, we explore and quantify their mobilization in patients with multiple myeloma undergoing chemotherapy upon administration of granulocyte colony-stimulating factor (G-CSF) plus/minus erythropoietin (EPO). PATIENTS AND METHODS: HPC were quantified by flow cytometry and functional assays within the blood of healthy donors and myeloma patients before and after chemotherapy followed by G-CSF or G-CSF + EPO given subcutaneously. The mRNA expression was studied by quantitative polymerase chain reaction (PCR). Cytokines and peripheral blood protease levels were measured by an enzyme-linked immunosorbent assay. RESULTS: EPO did not significantly alter the number of HPC mobilized by G-CSF alone, and mRNA specific for liver, brain, muscle and kidney was detected in both treatment groups. Quantitative PCR analysis revealed a 2.7-fold increased expression of glial fibrillary acidic protein after G-CSF + EPO administration compared to G-CSF alone (P = 0.003). The concentration of G-CSF rose from 62 +/- 22 pg/mL and 48 +/- 10 pg/mL to 28 +/- 9 ng/mL and 85 +/- 10 ng/mL after 10 d of treatment with G-CSF and G-CSF + EPO, respectively. The concentration of neutrophil elastase (NE) rose only in the G-CSF group by a factor 1.5. CONCLUSION: The alteration of G-CSF and NE levels as well as the expression of tissue committed RNA after the administration of EPO in addition to G-CSF indicate that different growth factors mobilize different stem cells that might potentially be used for the support of tissue repair in future treatment protocols.


Subject(s)
Erythropoietin/administration & dosage , Granulocyte Colony-Stimulating Factor/administration & dosage , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cells/cytology , Multiple Myeloma/therapy , RNA, Messenger/analysis , Antigens, CD34 , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Blood Cells , Case-Control Studies , Cell Count , Cytokines/blood , Erythropoietin/pharmacology , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/drug effects , Humans , Leukocyte Elastase/blood , Organ Specificity , RNA, Messenger/drug effects
14.
BMC Dev Biol ; 7: 12, 2007 Mar 02.
Article in English | MEDLINE | ID: mdl-17335568

ABSTRACT

BACKGROUND: The mechanisms responsible for the maintenance of pluripotency in human embryonic stem cells, and those that drive their commitment into particular differentiation lineages, are poorly understood. In fact, even our knowledge of the phenotype of hESC is limited, because the immunological and molecular criteria presently used to define this phenotype describe the properties of a heterogeneous population of cells. RESULTS: We used a novel approach combining immunological and transcriptional analysis (immunotranscriptional profiling) to compare gene expression in hESC populations at very early stages of differentiation. Immunotranscriptional profiling enabled us to identify novel markers of stem cells and their differentiated progeny, as well as novel potential regulators of hESC commitment and differentiation. The data show clearly that genes associated with the pluripotent state are downregulated in a coordinated fashion, and that they are co-expressed with lineage specific transcription factors in a continuum during the early stages of stem cell differentiation. CONCLUSION: These findings, that show that maintenance of pluripotency and lineage commitment are dynamic, interactive processes in hESC cultures, have important practical implications for propagation and directed differentiation of these cells, and for the interpretation of mechanistic studies of hESC renewal and commitment. Since embryonic stem cells at defined stages of commitment can be isolated in large numbers by immunological means, they provide a powerful model for studying molecular genetics of stem cell commitment in the embryo.


Subject(s)
Embryonic Stem Cells/cytology , Gene Expression Profiling , Oligonucleotide Array Sequence Analysis , Pluripotent Stem Cells/cytology , Animals , Cell Culture Techniques , Cell Line , Cell Lineage , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation, Developmental , Genetic Markers , Humans , Mice , Recombinant Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sialoglycoproteins/genetics
15.
Acta Biomater ; 52: 145-158, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28017869

ABSTRACT

Tissue engineering technology platforms constitute a unique opportunity to integrate cells and extracellular matrix (ECM) proteins into scaffolds and matrices that mimic the natural microenvironment in vitro. The development of tissue-engineered 3D models that mimic the endosteal microenvironment enables researchers to discover the causes and improve treatments for blood and immune-related diseases. The aim of this study was to establish a physiologically relevant in vitro model using 3D printed scaffolds to assess the contribution of human cells to the formation of a construct that mimics human endosteum. Melt electrospun written scaffolds were used to compare the suitability of primary human osteoblasts (hOBs) and placenta-derived mesenchymal stem cells (plMSCs) in (non-)osteogenic conditions and with different surface treatments. Using osteogenic conditions, hOBs secreted a dense ECM with enhanced deposition of endosteal proteins, such as fibronectin and vitronectin, and osteogenic markers, such as osteopontin and alkaline phosphatase, compared to plMSCs. The expression patterns of these proteins were reproducibly identified in hOBs derived from three individual donors. Calcium phosphate-coated scaffolds induced the expression of osteocalcin by hOBs when maintained in osteogenic conditions. The tissue-engineered endosteal microenvironment supported the growth and migration of primary human haematopoietic stem cells (HSCs) when compared to HSCs maintained using tissue culture plastic. This 3D testing platform represents an endosteal bone-like tissue and warrants future investigation for the maintenance and expansion of human HSCs. STATEMENT OF SIGNIFICANCE: This work is motivated by the recent interest in melt electrospinning writing, a 3D printing technique used to produce porous scaffolds for biomedical applications in regenerative medicine. Our team has been among the pioneers in building a new class of melt electrospinning devices for scaffold-based tissue engineering. These scaffolds allow structural support for various cell types to invade and deposit their own ECM, mimicking a characteristic 3D microenvironment for experimental studies. We used melt electrospun written polycaprolactone scaffolds to develop an endosteal bone-like tissue that promotes the growth of HSCs. We combine tissue engineering concepts with cell biology and stem cell research to design a physiologically relevant niche that is of prime interest to the scientific community.


Subject(s)
Biomimetic Materials/chemical synthesis , Bone Substitutes/chemical synthesis , Electroplating/methods , Extracellular Matrix/chemistry , Hematopoietic Stem Cells/cytology , Tissue Scaffolds , Cell Proliferation , Cell Survival , Cells, Cultured , Equipment Design , Female , Hematopoietic Stem Cells/physiology , Hot Temperature , Humans , Male , Printing, Three-Dimensional , Tissue Engineering/instrumentation , Tissue Engineering/methods
16.
Mater Sci Eng C Mater Biol Appl ; 79: 363-371, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28629029

ABSTRACT

Mesenchymal stem cells (MSCs) have the potential to revolutionize medicine due to their ability to differentiate into specific lineages for targeted tissue repair. Development of materials and cell culture platforms that improve differentiation of either autologous or allogenic stem cell sources into specific lineages would enhance clinical utilization of MCSs. In this study, nanoscale amyloid fibrils were evaluated as substrate materials to encourage viability, proliferation, multipotency, and differentiation of MSCs. Fibrils assembled from the proteins lysozyme or ß-lactoglobulin, with and without chitosan coatings, were deposited on planar mica surfaces. MSCs were cultured and differentiated on fibril-covered surfaces, as well as on unstructured controls and tissue culture plastic. Expression of CD44 and CD90 proteins indicated that multipotency was maintained for all fibrils, and osteogenic differentiation was similarly comparable among all tested materials. MSCs grown for 7days on fibril-covered surfaces favored multicellular spheroid formation and demonstrated a >75% increase in adipogenesis compared to tissue culture plastic controls, although this benefit could only be achieved if MSCs were transferred to TCP for the final differentiation step. The largest spheroids and greatest tendency to undergo adipogenesis was evidenced among MSCs grown on fibrils coated with the positively-charged polysaccharide chitosan, suggesting that spheroid formation is prompted by both topography and cell-surface interactivity and that there is a connection between multicellular spheroid formation and adipogenesis.


Subject(s)
Mesenchymal Stem Cells , Adipogenesis , Amyloid , Cell Differentiation , Cells, Cultured , Chitosan , Humans , Osteogenesis
17.
Int J Cardiol ; 109(1): 21-7, 2006 Apr 28.
Article in English | MEDLINE | ID: mdl-15970342

ABSTRACT

Current stem cell protocols for ischemic heart disease are limited by the small numbers of cells that can be obtained by bone marrow aspirate. To increase myocardial delivery of bone marrow stem cells in patients with chronic ischemic heart disease (CIHD), we used granulocyte colony stimulating factor (G-CSF) for bone marrow mobilization of CD34+ cells, enabling intracoronary infusion of large numbers of CD34+ stem cells. Patients with CIHD (n = 5) demonstrated significantly reduced numbers of CD34+ cells mobilized by G-CSF in comparison to age-matched controls. Sustained reduction in anginal symptoms and improvement in quality of life scores was seen in all patients following infusion of cells. Moreover, mean collateral flow grade at 12-month follow-up angiography significantly improved, indicating sustained myocardial neovascularization. No proliferative retinopathy was induced and no in-stent restenosis seen. However, in two patients with documented increase in collateral flow, complications arose, one developing an acute coronary syndrome and the other a lentigo maligna. These results demonstrate the feasibility of G-CSF mobilization, leukapheresis and intracoronary transfer of CD34+ stem cells in patients with CIHD, but longer-term studies are required to ensure that this protocol is safe and effective.


Subject(s)
Antigens, CD34/administration & dosage , Granulocyte Colony-Stimulating Factor/therapeutic use , Hematopoietic Stem Cell Mobilization , Myocardial Ischemia/therapy , Aged , Angioplasty, Balloon, Coronary , Collateral Circulation , Coronary Angiography , Female , Follow-Up Studies , Humans , Leukapheresis , Male , Middle Aged , Quality of Life , Tomography, Emission-Computed, Single-Photon , Ventricular Function, Left , Ventricular Remodeling
18.
Nat Biotechnol ; 34(11): 1168-1179, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27748754

ABSTRACT

The ability to generate hematopoietic stem cells from human pluripotent cells would enable many biomedical applications. We find that hematopoietic CD34+ cells in spin embryoid bodies derived from human embryonic stem cells (hESCs) lack HOXA expression compared with repopulation-competent human cord blood CD34+ cells, indicating incorrect mesoderm patterning. Using reporter hESC lines to track the endothelial (SOX17) to hematopoietic (RUNX1C) transition that occurs in development, we show that simultaneous modulation of WNT and ACTIVIN signaling yields CD34+ hematopoietic cells with HOXA expression that more closely resembles that of cord blood. The cultures generate a network of aorta-like SOX17+ vessels from which RUNX1C+ blood cells emerge, similar to hematopoiesis in the aorta-gonad-mesonephros (AGM). Nascent CD34+ hematopoietic cells and corresponding cells sorted from human AGM show similar expression of cell surface receptors, signaling molecules and transcription factors. Our findings provide an approach to mimic in vitro a key early stage in human hematopoiesis for the generation of AGM-derived hematopoietic lineages from hESCs.


Subject(s)
Embryonic Stem Cells/cytology , Hematopoietic Stem Cells/cytology , Homeodomain Proteins/metabolism , Mesonephros/cytology , Mesonephros/embryology , Neovascularization, Physiologic/physiology , Aorta/cytology , Aorta/embryology , Aorta/growth & development , Cell Differentiation/physiology , Cells, Cultured , Embryonic Stem Cells/physiology , Gonads/cytology , Gonads/embryology , Gonads/growth & development , Hematopoietic Stem Cells/physiology , Humans , Mesonephros/growth & development
19.
Stem Cell Res Ther ; 6: 58, 2015 Apr 16.
Article in English | MEDLINE | ID: mdl-25888759

ABSTRACT

INTRODUCTION: Recent studies in the literature have highlighted the critical role played by cell signalling in determining haemopoietic stem cell (HSC) fate within ex vivo culture systems. Stimulatory signals can enhance proliferation and promote differentiation, whilst inhibitory signals can significantly limit culture output. METHODS: Numerical models of various mitigation strategies are presented and applied to determine effectiveness of these strategies toward mitigation of paracrine inhibitory signalling inherent in these culture systems. The strategies assessed include mixing, media-exchange, fed-batch and perfusion. RESULTS: The models predict that significant spatial concentration gradients exist in typical cell cultures, with important consequences for subsequent cell expansion. Media exchange is shown to be the most effective mitigation strategy, but remains labour intensive and difficult to scale-up for large culture systems. The fed-batch strategy is only effective at very small Peclet number, and its effect is diminished as the cell culture volume grows. Conversely, mixing is effective at high Peclet number, and ineffective at low Peclet number. The models predict that cell expansion in fed-batch cultures becomes independent of increasing dilution rate, consistent with experimental results previously reported in the literature. In contrast, the models predict that increasing the flow rate in perfused cultures will lead to increased cell expansion, indicating the suitability of perfusion for use as an automated, tunable strategy. The effect of initial cell seeding density is also investigated, with the model showing that perfusion outperforms dilution for all densities considered. CONCLUSIONS: The models predict that the impact of inhibitory signalling in HSC cultures can be mitigated against using media manipulation strategies, with the optimal strategy dependent upon the protein diffusion time-scale relative to the media manipulation time-scale. The key messages from this study can be applied to any complex cell culture scenario where cell-cell interactions and paracrine signalling networks impact upon cell fate and cell expansion.


Subject(s)
Hematopoietic Stem Cells/cytology , Models, Biological , Paracrine Communication , Animals , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Paracrine Communication/drug effects , Transforming Growth Factor beta/pharmacology
20.
Tissue Eng Part B Rev ; 21(2): 187-202, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25341031

ABSTRACT

Melt electrospinning is an emerging fiber-based manufacturing technique that can be used to design and build scaffolds suitable for many tissue engineering (TE) applications. Contrary to the widely used solution electrospinning, the melt process is solvent-free and therefore volatility and toxicity issues associated with solvents can be avoided. Furthermore, molten polymers are often viscous and nonconductive, making them candidates for generating electrospinning jets without electrical instabilities. This in turn permits a precise and predictable fiber deposition in the combination with moving collectors, termed melt electrospinning writing (MEW), allows the layer-by-layer fabrication of small to large volume scaffolds with specific designs, shapes and thicknesses. In vitro studies have demonstrated that scaffolds designed and fabricated via MEW can support cell attachment, proliferation and extracellular matrix formation, as well as cell infiltration throughout the thickness of the scaffold facilitated by the large pores and pore interconnectivity. Moreover, in vivo studies show that scaffolds designed for specific tissue regeneration strategies performed superbly. This review describes the state-of-the-art and unique perspectives of melt electrospinning and its writing applied to scaffold-based TE.


Subject(s)
Biocompatible Materials , Tissue Engineering/methods , Tissue Scaffolds , Animals , Humans , Tissue Engineering/instrumentation
SELECTION OF CITATIONS
SEARCH DETAIL