Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 107
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Mol Sci ; 24(17)2023 Aug 26.
Article in English | MEDLINE | ID: mdl-37686058

ABSTRACT

Adoptive transfer of cultured BMSCs was shown to be immune-suppressive in various inflammatory settings. Many factors play a role in the process, but no master regulator of BMSC-driven immunomodulation was identified. Consequently, an assay that might predict BMSC product efficacy is still unavailable. Below, we show that BMSC donor variability can be monitored by IL-10 production of monocytes/macrophages using THP-1 cells (immortalized monocytic leukemia cells) co-cultured with BMSCs. Using a mixed lymphocyte reaction (MLR) assay, we also compared the ability of the different donor BMSCs to suppress T-cell proliferation, another measure of their immune-suppressive ability. We found that the BMSCs from a donor that induced the most IL-10 production were also the most efficient in suppressing T-cell proliferation. Transcriptome studies showed that the most potent BMSC batch also had higher expression of several known key immunomodulatory molecules such as hepatocyte growth factor (HGF), PDL1, and numerous members of the PGE2 pathway, including PTGS1 and TLR4. Multiplex ELISA experiments revealed higher expression of HGF and IL6 by the most potent BMSC donor. Based on these findings, we propose that THP-1 cells may be used to assess BMSC immunosuppressive activity as a product characterization assay.


Subject(s)
Bone Marrow , Leukemia, Monocytic, Acute , Humans , Pilot Projects , Interleukin-10 , Cell Line , Stromal Cells
2.
Am J Hum Genet ; 104(5): 925-935, 2019 05 02.
Article in English | MEDLINE | ID: mdl-30982609

ABSTRACT

Colony stimulating factor 1 receptor (CSF1R) plays key roles in regulating development and function of the monocyte/macrophage lineage, including microglia and osteoclasts. Mono-allelic mutations of CSF1R are known to cause hereditary diffuse leukoencephalopathy with spheroids (HDLS), an adult-onset progressive neurodegenerative disorder. Here, we report seven affected individuals from three unrelated families who had bi-allelic CSF1R mutations. In addition to early-onset HDLS-like neurological disorders, they had brain malformations and skeletal dysplasia compatible to dysosteosclerosis (DOS) or Pyle disease. We identified five CSF1R mutations that were homozygous or compound heterozygous in these affected individuals. Two of them were deep intronic mutations resulting in abnormal inclusion of intron sequences in the mRNA. Compared with Csf1r-null mice, the skeletal and neural phenotypes of the affected individuals appeared milder and variable, suggesting that at least one of the mutations in each affected individual is hypomorphic. Our results characterized a unique human skeletal phenotype caused by CSF1R deficiency and implied that bi-allelic CSF1R mutations cause a spectrum of neurological and skeletal disorders, probably depending on the residual CSF1R function.


Subject(s)
Brain/abnormalities , Leukoencephalopathies/etiology , Mutation , Osteochondrodysplasias/etiology , Osteosclerosis/etiology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Adolescent , Adult , Alleles , Animals , Brain/metabolism , Brain/pathology , Child, Preschool , Female , Humans , Leukoencephalopathies/pathology , Male , Mice , Mice, Knockout , Osteochondrodysplasias/pathology , Osteosclerosis/pathology , Phenotype , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Young Adult
3.
Stem Cells ; 38(9): 1107-1123, 2020 09.
Article in English | MEDLINE | ID: mdl-32442326

ABSTRACT

Human pluripotent stem cells (hPSCs) can provide a platform to model bone organogenesis and disease. To reflect the developmental process of the human skeleton, hPSC differentiation methods should include osteogenic progenitors (OPs) arising from three distinct embryonic lineages: the paraxial mesoderm, lateral plate mesoderm, and neural crest. Although OP differentiation protocols have been developed, the lineage from which they are derived, as well as characterization of their genetic and molecular differences, has not been well reported. Therefore, to generate lineage-specific OPs from human embryonic stem cells and human induced pluripotent stem cells, we employed stepwise differentiation of paraxial mesoderm-like cells, lateral plate mesoderm-like cells, and neural crest-like cells toward their respective OP subpopulation. Successful differentiation, confirmed through gene expression and in vivo assays, permitted the identification of transcriptomic signatures of all three cell populations. We also report, for the first time, high FGF1 levels in neural crest-derived OPs-a notable finding given the critical role of fibroblast growth factors (FGFs) in osteogenesis and mineral homeostasis. Our results indicate that FGF1 influences RUNX2 levels, with concomitant changes in ERK1/2 signaling. Overall, our study further validates hPSCs' power to model bone development and disease and reveals new, potentially important pathways influencing these processes.


Subject(s)
Cell Differentiation , Cell Lineage , Core Binding Factor Alpha 1 Subunit/metabolism , Fibroblast Growth Factor 1/metabolism , Neural Crest/cytology , Osteogenesis , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Animals , Humans , MAP Kinase Signaling System , Male , Mice , Principal Component Analysis , Transcriptome/genetics
4.
Int J Mol Sci ; 22(8)2021 Apr 13.
Article in English | MEDLINE | ID: mdl-33924333

ABSTRACT

Bone formation starts near the end of the embryonic stage of development and continues throughout life during bone modeling and growth, remodeling, and when needed, regeneration. Bone-forming cells, traditionally termed osteoblasts, produce, assemble, and control the mineralization of the type I collagen-enriched bone matrix while participating in the regulation of other cell processes, such as osteoclastogenesis, and metabolic activities, such as phosphate homeostasis. Osteoblasts are generated by different cohorts of skeletal stem cells that arise from different embryonic specifications, which operate in the pre-natal and/or adult skeleton under the control of multiple regulators. In this review, we briefly define the cellular identity and function of osteoblasts and discuss the main populations of osteoprogenitor cells identified to date. We also provide examples of long-known and recently recognized regulatory pathways and mechanisms involved in the specification of the osteogenic lineage, as assessed by studies on mice models and human genetic skeletal diseases.


Subject(s)
Osteoblasts/cytology , Osteogenesis , Stem Cells/cytology , Animals , Bone and Bones/cytology , Bone and Bones/embryology , Epigenesis, Genetic , Humans , Osteogenesis/genetics , Signal Transduction
5.
Development ; 144(6): 1035-1044, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28292847

ABSTRACT

Human umbilical cord blood (CB) has attracted much attention as a reservoir for functional hematopoietic stem and progenitor cells, and, recently, as a source of blood-borne fibroblasts (CB-BFs). Previously, we demonstrated that bone marrow stromal cell (BMSC) and CB-BF pellet cultures make cartilage in vitro Furthermore, upon in vivo transplantation, BMSC pellets remodelled into miniature bone/marrow organoids. Using this in vivo model, we asked whether CB-BF populations that express characteristics of the hematopoietic stem cell (HSC) niche contain precursors that reform the niche. CB ossicles were regularly observed upon transplantation. Compared with BM ossicles, CB ossicles showed a predominance of red marrow over yellow marrow, as demonstrated by histomorphological analyses and the number of hematopoietic cells isolated within ossicles. Marrow cavities from CB and BM ossicles included donor-derived CD146-expressing osteoprogenitors and host-derived mature hematopoietic cells, clonogenic lineage-committed progenitors and HSCs. Furthermore, human CD34+ cells transplanted into ossicle-bearing mice engrafted and maintained human HSCs in the niche. Our data indicate that CB-BFs are able to recapitulate the conditions by which the bone marrow microenvironment is formed and establish complete HSC niches, which are functionally supportive of hematopoietic tissue.


Subject(s)
Bone Marrow Cells/cytology , Fetal Blood/cytology , Fibroblasts/cytology , Hematopoietic Stem Cells/cytology , Organoids/cytology , Stem Cell Niche , Adult , Cell Compartmentation , Child , Fibroblasts/transplantation , Hematopoietic Stem Cell Transplantation , Homeostasis , Humans , Stem Cell Niche/genetics , Stromal Cells/cytology
6.
Stem Cells ; 36(1): 11-21, 2018 01.
Article in English | MEDLINE | ID: mdl-28948674

ABSTRACT

Lineage commitment and differentiation of skeletal stem cells/bone marrow stromal cells (SSCs/BMSCs, often called bone marrow-derived "mesenchymal stem/stromal" cells) offer an important opportunity to study skeletal and hematopoietic diseases, and for tissue engineering and regenerative medicine. Currently, many studies in this field have relied on cell lineage tracing methods in mouse models, which have provided a significant advancement in our knowledge of skeletal and hematopoietic stem-cell niches in bone marrow (BM). However, there is a lack of agreement in numerous fundamental areas, including origins of various BM stem-cell niches, cell identities, and their physiological roles in the BM. In order to resolve these issues, we propose a new hypothesis of "paralogous" stem-cell niches (PSNs); that is, progressively altered parallel niches within an individual species throughout the life span of the organism. A putative PSN code seems to be plausible based on analysis of transcriptional signatures in two representative genes that encode Nes-GFP and leptin receptors, which are frequently used to monitor SSC lineage development in BM. Furthermore, we suggest a dynamic paralogous BM niche (PBMN) model that elucidates the coupling and uncoupling mechanisms between BM stem-cell niches and their zones of active regeneration during different developmental stages. Elucidation of these PBMNs would enable us to resolve the existing controversies, thus paving the way to achieving precision regenerative medicine and pharmaceutical applications based on these BM cell resources. Stem Cells 2018;36:11-21.


Subject(s)
Bone Marrow/metabolism , Hematopoietic Stem Cells/metabolism , Stem Cell Niche/genetics , Stem Cells/metabolism , Cell Differentiation , Cell Lineage , Humans
8.
Development ; 142(6): 1023-7, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25758217

ABSTRACT

Skeletal stem cells (SSCs) reside in the postnatal bone marrow and give rise to cartilage, bone, hematopoiesis-supportive stroma and marrow adipocytes in defined in vivo assays. These lineages emerge in a specific sequence during embryonic development and post natal growth, and together comprise a continuous anatomical system, the bone-bone marrow organ. SSCs conjoin skeletal and hematopoietic physiology, and are a tool for understanding and ameliorating skeletal and hematopoietic disorders. Here and in the accompanying poster, we concisely discuss the biology of SSCs in the context of the development and postnatal physiology of skeletal lineages, to which their use in medicine must remain anchored.


Subject(s)
Bone and Bones/cytology , Cell Lineage/physiology , Mesenchymal Stem Cells/cytology , Multipotent Stem Cells/physiology , Stem Cell Niche , Adipocytes/cytology , Cartilage/cytology , Humans , Terminology as Topic
9.
J Transl Med ; 16(1): 65, 2018 03 14.
Article in English | MEDLINE | ID: mdl-29540180

ABSTRACT

BACKGROUND: Bone marrow stromal cells (BMSCs) have classically been cultured in media supplemented with fetal bovine serum (FBS). As an alternative to FBS, pooled solvent detergent apheresis platelets, HPGF-C18, was evaluated for BMSC culture. METHODS: A comparison of passage 2 BMSC growth revealed that 10% HPGF-C18 produced similar cell numbers as 20% FBS. Marrow aspirates from 5 healthy subjects were cultured for 4 passages in 10% HPGF-C18 or 20% FBS and were analyzed for proliferation, colony formation efficiency (CFE), surface marker expression, suppression of mixed lymphocyte reactions (MLRs), global gene and microRNA expression analysis. BMSC supernatant cytokine and growth factor concentrations were also compared. RESULTS: Primary cultures of marrow aspirates in 10% HPGF-C18 and 20% FBS yielded similar numbers and CFE. After 4 passages, 10% HPGF-C18 and 20% FBS yielded similar numbers of BMSCs, surface marker expression patterns and immunosuppression effects. Gene and microRNA expression analysis revealed that BMSCs cultured under the two conditions had distinct expression profiles. Gene Set Enrichment Analysis (GSEA) revealed HPGF-C18-cultured BMSCs were enriched in metabolic processing and biosynthetic pathways, cell proliferation and cell cycle pathways, and immune response pathways. FBS-cultured BMSCs were enriched in MAPK signaling, TGF-beta signaling, cell adhesion and extracellular matrix pathways. Differently expressed microRNAs were related to the osteogenesis of BMSCs. The supernatant of HPGF-C18 BMSCs had higher levels of PEDF and TGFB1 and lower levels of IL6, VEGF, SDF1 and PLGF. CONCLUSIONS: Traditional measures, expansion, surface marker expression and inhibition of MLRs suggest that BMSC cultured in HPGF-C18 and FBS were similar, but analysis at the molecular level revealed many differences. BMSCs cultured in HPGF-C18 should be assessed in specific functional assays that reflect application-specific potency before substituting FBS with HPGF-C18.


Subject(s)
Bone Marrow Cells/cytology , Cell Culture Techniques/methods , Platelet-Derived Growth Factor/pharmacology , Serum/metabolism , Animals , Biomarkers/metabolism , Bone Marrow Cells/drug effects , Cattle , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/metabolism , Gene Expression Regulation/drug effects , Humans , Lymphocyte Culture Test, Mixed , MicroRNAs/genetics , MicroRNAs/metabolism , Stromal Cells/cytology , Stromal Cells/drug effects , Transcriptome/genetics
10.
Cytotherapy ; 20(11): 1381-1400, 2018 11.
Article in English | MEDLINE | ID: mdl-30316562

ABSTRACT

The Signature Series Symposium "Cellular Therapies for Orthopaedics and Musculoskeletal Disease Proven and Unproven Therapies-Promise, Facts and Fantasy" was held as a pre-meeting of the 26th International Society for Cellular Therapy (ISCT) annual congress in Montreal, Canada, May 2, 2018. This was the first ISCT program that was entirely dedicated to the advancement of cell-based therapies for musculoskeletal diseases. Cellular therapies in musculoskeletal medicine are a source of great promise and opportunity. They are also the source of public controversy, confusion and misinformation. Patients, clinicians, scientists, industry and government share a commitment to clear communication and responsible development of the field. Therefore, this symposium convened thought leaders from around the world in a forum designed to catalyze communication and collaboration to bring the greatest possible innovation and value to patients with musculoskeletal conditions.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Musculoskeletal Diseases/therapy , Animals , Cell- and Tissue-Based Therapy/standards , Fantasy , Humans , Musculoskeletal Diseases/veterinary , Orthopedics , Regenerative Medicine/methods , Societies, Scientific , Translational Research, Biomedical/legislation & jurisprudence , Translational Research, Biomedical/standards , Veterinary Medicine/methods
11.
EMBO J ; 32(11): 1489-95, 2013 May 29.
Article in English | MEDLINE | ID: mdl-23644381

ABSTRACT

At the time of writing, the Italian Parliament is debating a new law that would make it legal to practice an unproven stem cell treatment in public hospitals. The treatment, offered by a private non-medical organization, may not be safe, lacks a rationale, and violates current national laws and European regulations. This case raises multiple concerns, most prominently the urgent need to protect patients who are severely ill, exposed to significant risks, and vulnerable to exploitation. The scientific community must consider the context-social, financial, medical, legal-in which stem cell science is currently situated and the need for stringent regulation. Additional concerns are emerging. These emanate from the novel climate, created within science itself, and stem cell science in particular, by the currently prevailing model of 'translational medicine'. Only rigorous science and rigorous regulation can ensure translation of science into effective therapies rather than into ineffective market products, and mark, at the same time, the sharp distinction between the striving for new therapies and the deceit of patients.


Subject(s)
Stem Cell Transplantation/legislation & jurisprudence , Europe , Humans , Italy , Patient Safety , Stem Cell Transplantation/adverse effects , Stem Cell Transplantation/standards , Stem Cells , Translational Research, Biomedical
12.
Blood ; 125(5): 793-802, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25499762

ABSTRACT

Dyskeratosis congenita (DC) is an inherited multisystem disorder, characterized by oral leukoplakia, nail dystrophy, and abnormal skin pigmentation, as well as high rates of bone marrow (BM) failure, solid tumors, and other medical problems such as osteopenia. DC and telomere biology disorders (collectively referred to as TBD here) are caused by germline mutations in telomere biology genes leading to very short telomeres and limited proliferative potential of hematopoietic stem cells. We found that skeletal stem cells (SSCs) within the BM stromal cell population (BMSCs, also known as BM-derived mesenchymal stem cells), may contribute to the hematologic phenotype. TBD-BMSCs exhibited reduced clonogenicity, spontaneous differentiation into adipocytes and fibrotic cells, and increased senescence in vitro. Upon in vivo transplantation into mice, TBD-BMSCs failed to form bone or support hematopoiesis, unlike normal BMSCs. TERC reduction (a TBD-associated gene) in normal BMSCs by small interfering TERC-RNA (siTERC-RNA) recapitulated the TBD-BMSC phenotype by reducing proliferation and secondary colony-forming efficiency, and by accelerating senescence in vitro. Microarray profiles of control and siTERC-BMSCs showed decreased hematopoietic factors at the messenger RNA level and decreased secretion of factors at the protein level. These findings are consistent with defects in SSCs/BMSCs contributing to BM failure in TBD.


Subject(s)
Bone Marrow Cells/metabolism , Dyskeratosis Congenita/genetics , Mesenchymal Stem Cells/metabolism , RNA/genetics , Telomerase/genetics , Telomere/metabolism , Adolescent , Adult , Animals , Base Sequence , Bone Marrow Cells/pathology , Cell Differentiation , Cell Proliferation , Cellular Senescence , Child , Child, Preschool , Colony-Forming Units Assay , DNA Helicases/genetics , DNA Helicases/metabolism , Dyskeratosis Congenita/pathology , Female , Hematopoiesis/genetics , Humans , Male , Mesenchymal Stem Cells/pathology , Mice , Middle Aged , Molecular Sequence Data , Mutation , RNA/antagonists & inhibitors , RNA/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Telomerase/antagonists & inhibitors , Telomerase/metabolism , Telomere/chemistry , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism
13.
Skeletal Radiol ; 46(10): 1435-1439, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28660402

ABSTRACT

Bisphosphonates (BPs) are currently used in the treatment of diverse bone diseases including fibrous dysplasia of bone (FD). In pediatric patients, a radiographic consequence of cyclical administration of BPs is the development of apo-, epi-, and meta-physeal sclerotic bands, otherwise known as zebra lines, which result from the temporary inhibition of osteoclastic activity at the time of drug treatment. We report here on a child with McCune-Albright syndrome (FD in addition to hyperfunctioning endocrinopathies and skin hyperpigmentation) treated with cyclical intravenous infusions of pamidronate in which conventional radiography, contact microradiography, histology, and backscattered electron image analysis demonstrated that zebra lines formed only where bone was normal, were arrested at the boundary between FD-unaffected and FD-affected bone where bone is sclerotic, and were absent within the undermineralized FD bone. Moreover, in spite of the treatment, the FD lesions continued to expand. This case report is unique because no previously published studies correlated the radiographic and the histologic features of BP-induced zebra lines in the metaphysis of an FD-affected long bone of the limbs.


Subject(s)
Diphosphonates/adverse effects , Femur/drug effects , Femur/diagnostic imaging , Fibrous Dysplasia, Polyostotic/diagnostic imaging , Fibrous Dysplasia, Polyostotic/drug therapy , Adolescent , Disease Progression , Female , Fibrous Dysplasia, Polyostotic/surgery , Humans , Pamidronate
14.
J Biol Chem ; 290(22): 14004-18, 2015 May 29.
Article in English | MEDLINE | ID: mdl-25864198

ABSTRACT

WISP1/CCN4 (hereafter referred to as WISP1), a member of the CCN family, is found in mineralized tissues and is produced by osteoblasts and their precursors. In this study, Wisp1-deficient (Wisp1(-/-)) mice were generated. Using dual-energy x-ray absorptiometry, we showed that by 3 months, the total bone mineral density of Wisp1(-/-) mice was significantly lower than that of WT mice. Further investigation by micro-computed tomography showed that female Wisp1(-/-) mice had decreased trabecular bone volume/total volume and that both male and female Wisp1(-/-) mice had decreased cortical bone thickness accompanied by diminished biomechanical strength. The molecular basis for decreased bone mass in Wisp1(-/-) mice arises from reduced bone formation likely caused by osteogenic progenitors that differentiate poorly compared with WT cells. Osteoclast precursors from Wisp1(-/-) mice developed more tartrate-resistant acid phosphatase-positive cells in vitro and in transplants, suggesting that WISP1 is also a negative regulator of osteoclast differentiation. When bone turnover (formation and resorption) was induced by ovariectomy, Wisp1(-/-) mice had lower bone mineral density compared WT mice, confirming the potential for multiple roles for WISP1 in controlling bone homeostasis. Wisp1(-/-) bone marrow stromal cells had reduced expression of ß-catenin and its target genes, potentially caused by WISP1 inhibition of SOST binding to LRP6. Taken together, our data suggest that the decreased bone mass found in Wisp1(-/-) mice could potentially be caused by an insufficiency in the osteodifferentiation capacity of bone marrow stromal cells arising from diminished Wnt signaling, ultimately leading to altered bone turnover and weaker biomechanically compromised bones.


Subject(s)
Bone Remodeling , Bone and Bones/metabolism , CCN Intercellular Signaling Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Wnt Signaling Pathway , Alleles , Animals , Bone Marrow Cells/cytology , Cell Differentiation , Cells, Cultured , Extracellular Matrix/metabolism , Female , Low Density Lipoprotein Receptor-Related Protein-1 , Male , Mice , Mice, Knockout , Osteoblasts/metabolism , Osteoclasts/metabolism , RNA, Messenger/metabolism , Receptors, LDL/metabolism , Recombination, Genetic , Stromal Cells/cytology , Tumor Suppressor Proteins/metabolism , X-Ray Microtomography
15.
Stem Cells ; 33(4): 1304-19, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25524638

ABSTRACT

The tumor suppressor, p53, plays a critical role in suppressing osteosarcoma. Bone marrow stromal cells (BMSCs, also known as bone marrow-derived mesenchymal stem cells) have been suggested to give rise to osteosarcomas. However, the role of p53 in BMSCs has not been extensively explored. Here, we report that p53 regulates the lineage choice of mouse BMSCs (mBMSCs). Compared to mBMSCs with wild-type p53, mBMSCs deficient in p53 have enhanced osteogenic differentiation, but with similar adipogenic and chondrogenic differentiation. The role of p53 in inhibiting osteogenic lineage differentiation is mainly through the action of Runx2, a master transcription factor required for the osteogenic differentiation of mBMSCs. We find that p53 indirectly represses the expression of Runx2 by activating the microRNA-34 family, which suppresses the translation of Runx2. Since osteosarcoma may derive from BMSCs, we examined whether p53 has a role in the osteogenic differentiation of osteosarcoma cells and found that osteosarcoma cells with p53 deletion have higher levels of Runx2 and faster osteogenic differentiation than those with wild-type p53. A systems biology approach reveals that p53-deficient mBMSCs are more closely related to human osteosarcoma while mBMSCs with wild-type p53 are similar to normal human BMSCs. In summary, our results indicate that p53 activity can influence cell fate specification of mBMSCs, and provide molecular and cellular insights into the observation that p53 loss is associated with increased osteosarcoma incidence.


Subject(s)
Cell Differentiation/physiology , Mesenchymal Stem Cells/metabolism , Osteogenesis/physiology , Tumor Suppressor Protein p53/deficiency , Animals , Cell Line, Tumor , Cells, Cultured , Humans , Mice , Mice, Knockout
16.
Stem Cells ; 33(4): 1200-12, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25532725

ABSTRACT

The use of bone marrow-derived mesenchymal stromal cells (BMSC) in the treatment of alloimmune and autoimmune conditions has generated much interest, yet an understanding of the therapeutic mechanism remains elusive. We therefore explored immune modulation by a clinical-grade BMSC product in a model of human-into-mouse xenogeneic graft-versus-host disease (x-GVHD) mediated by human CD4(+) Th1 cells. BMSC reversed established, lethal x-GVHD through marked inhibition of Th1 cell effector function. Gene marking studies indicated BMSC engraftment was limited to the lung; furthermore, there was no increase in regulatory T cells, thereby suggesting a paracrine mechanism of BMSC action. BMSC recipients had increased serum CD73 expressing exosomes that promoted adenosine accumulation ex vivo. Importantly, immune modulation mediated by BMSC was fully abrogated by pharmacologic therapy with an adenosine A2A receptor antagonist. To investigate the potential clinical relevance of these mechanistic findings, patient serum samples collected pre- and post-BMSC treatment were studied for exosome content: CD73 expressing exosomes promoting adenosine accumulation were detected in post-BMSC samples. In conclusion, BMSC effectively modulate experimental GVHD through a paracrine mechanism that promotes adenosine-based immune suppression.


Subject(s)
Adenosine A2 Receptor Antagonists/pharmacology , Mesenchymal Stem Cells/immunology , Signal Transduction/immunology , Th1 Cells/immunology , Animals , Bone Marrow/drug effects , Bone Marrow/immunology , Coculture Techniques , Graft vs Host Disease/immunology , Humans , Mesenchymal Stem Cells/drug effects , Mice , Mice, Inbred NOD , Mice, SCID , Signal Transduction/drug effects , Th1 Cells/drug effects
17.
Stem Cells ; 32(5): 1278-88, 2014 May.
Article in English | MEDLINE | ID: mdl-24452962

ABSTRACT

Bone marrow mesenchymal stromal cells (BMSCs) have been used to treat acute graft-versus-host disease (GVHD) and other complications following allogeneic hematopoietic stem cell transplantation (SCT). We conducted a phase I trial using third party, early passage BMSCs for patients with steroid-refractory GVHD, tissue injury, or marrow failure following SCT to investigate safety and efficacy. To identify mechanisms of BMSC immunomodulation and tissue repair, patients were serially monitored for plasma GVHD biomarkers, cytokines, and lymphocyte phenotype. Ten subjects were infused a fixed dose of 2 × 10(6) BMSCs/kg intravenously weekly for three doses. There was no treatment-related toxicity (primary endpoint). Eight subjects were evaluable for response at 4 weeks after the last infusion. Five of the seven patients with steroid-refractory acute GVHD achieved a complete response, two of two patients with tissue injury (pneumomediastinum/pneumothorax) achieved resolution but there was no response in two subjects with delayed marrow failure. Rapid reductions in inflammatory cytokines were observed. Clinical responses correlated with a fall in biomarkers (Reg 3α, CK18, and Elafin) relevant for the site of GVHD or tissue injury. The GVHD complete responders survived significantly longer and had higher baseline absolute lymphocyte and central memory CD4 and CD8 counts. Cytokine changes also segregated with survival. These results confirm that BMSCs are associated with rapid clinical and biomarker responses in GVHD and tissue injury. However, BMSCs were ineffective in patients with prolonged GVHD with lower lymphocyte counts, which suggest that effective GVHD control by BMSCs requires a relatively intact immune system.


Subject(s)
Bone Marrow Cells/cytology , Graft vs Host Disease/therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Adult , Aged , Antigens, Neoplasm/blood , Biomarkers, Tumor/blood , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cytokines/blood , Elafin/blood , Female , Graft vs Host Disease/blood , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/methods , Humans , Infusions, Intravenous , Keratin-18/blood , Lectins, C-Type/blood , Lymphocyte Count , Male , Mediastinal Emphysema/blood , Mediastinal Emphysema/etiology , Mediastinal Emphysema/therapy , Middle Aged , Pancreatitis-Associated Proteins , Pneumothorax/blood , Pneumothorax/etiology , Pneumothorax/therapy , Survival Analysis , Transplantation, Homologous , Treatment Outcome , Young Adult
18.
Cytotherapy ; 17(7): 897-911, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25882666

ABSTRACT

BACKGROUND AIMS: Ex vivo expansion and serial passage of human bone marrow stromal cells (BMSCs, also known as bone marrow-derived mesenchymal stem cells) is required to obtain sufficient quantities for clinical therapy. The BMSC confluence criteria used to determine passage and harvest timing vary widely, and the impact of confluence on BMSC properties remains controversial. The effects of confluence on BMSC properties were studied and confluence-associated markers were identified. METHODS: BMSC characteristics were analyzed as they grew from 50% to 100% confluence, including viability, population doubling time, apoptosis, colony formation, immunosuppression, surface marker expression, global gene expression and microRNA expression. In addition, culture supernatant protein, glucose, lactate and pH levels were analyzed. RESULTS: Confluence-dependent changes were detected in the expression of several cell surface markers: 39 culture supernatant proteins, 26 microRNAs and 2078 genes. Many of these surface markers, proteins, microRNAs and genes have been reported to be important in BMSC function. The pigment epithelium-derived factor/vascular endothelial growth factor ratio increased with confluence, but 80% and 100% confluent BMSCs demonstrated a similar level of immunosuppression of mixed lymphocyte reactions. In addition, changes in lactate and glucose levels correlated with BMSC density. CONCLUSIONS: BMSC characteristics change as confluence increases. 100% confluent BMSCs may have compromised pro-angiogenesis properties but may retain their immunomodulatory properties. Supernatant lactate and glucose levels can be used to estimate confluence and ensure consistency in passage and harvest timing. Flow cytometry or microRNA expression can be used to confirm that the BMSCs have been harvested at the appropriate confluence.


Subject(s)
Bone Marrow Cells/cytology , Cell Culture Techniques , Cell Proliferation/physiology , Mesenchymal Stem Cells/cytology , Apoptosis/physiology , Biomarkers/metabolism , Cell- and Tissue-Based Therapy/methods , Cells, Cultured , Eye Proteins/metabolism , Flow Cytometry , Gene Expression , Gene Expression Profiling , Glucose/metabolism , Humans , Lactic Acid/metabolism , Male , Membrane Proteins/metabolism , MicroRNAs/biosynthesis , MicroRNAs/genetics , Nerve Growth Factors/metabolism , Serpins/metabolism , Vascular Endothelial Growth Factor A/metabolism
19.
Proc Natl Acad Sci U S A ; 109(18): 6927-32, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22509028

ABSTRACT

The bromodomain protein, BRD4, has been identified recently as a therapeutic target in acute myeloid leukemia, multiple myeloma, Burkitt's lymphoma, NUT midline carcinoma, colon cancer, and inflammatory disease; its loss is a prognostic signature for metastatic breast cancer. BRD4 also contributes to regulation of both cell cycle and transcription of oncogenes, HIV, and human papilloma virus (HPV). Despite its role in a broad range of biological processes, the precise molecular mechanism of BRD4 function remains unknown. We report that BRD4 is an atypical kinase that binds to the carboxyl-terminal domain (CTD) of RNA polymerase II and directly phosphorylates its serine 2 (Ser2) sites both in vitro and in vivo under conditions where other CTD kinases are inactive. Phosphorylation of the CTD Ser2 is inhibited in vivo by a BRD4 inhibitor that blocks its binding to chromatin. Our finding that BRD4 is an RNA polymerase II CTD Ser2 kinase implicates it as a regulator of eukaryotic transcription.


Subject(s)
Nuclear Proteins/metabolism , RNA Polymerase II/chemistry , RNA Polymerase II/metabolism , Transcription Factors/metabolism , Amino Acid Substitution , Animals , Binding Sites/genetics , Cell Cycle Proteins , Cells, Cultured , Humans , Mice , Mutagenesis, Site-Directed , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Phosphorylation , Protein Structure, Tertiary , RNA Polymerase II/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine/chemistry , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription, Genetic
20.
N Engl J Med ; 365(7): 611-9, 2011 Aug 18.
Article in English | MEDLINE | ID: mdl-21793738

ABSTRACT

BACKGROUND: The Proteus syndrome is characterized by the overgrowth of skin, connective tissue, brain, and other tissues. It has been hypothesized that the syndrome is caused by somatic mosaicism for a mutation that is lethal in the nonmosaic state. METHODS: We performed exome sequencing of DNA from biopsy samples obtained from patients with the Proteus syndrome and compared the resultant DNA sequences with those of unaffected tissues obtained from the same patients. We confirmed and extended an observed association, using a custom restriction-enzyme assay to analyze the DNA in 158 samples from 29 patients with the Proteus syndrome. We then assayed activation of the AKT protein in affected tissues, using phosphorylation-specific antibodies on Western blots. RESULTS: Of 29 patients with the Proteus syndrome, 26 had a somatic activating mutation (c.49G→A, p.Glu17Lys) in the oncogene AKT1, encoding the AKT1 kinase, an enzyme known to mediate processes such as cell proliferation and apoptosis. Tissues and cell lines from patients with the Proteus syndrome harbored admixtures of mutant alleles that ranged from 1% to approximately 50%. Mutant cell lines showed greater AKT phosphorylation than did control cell lines. A pair of single-cell clones that were established from the same starting culture and differed with respect to their mutation status had different levels of AKT phosphorylation. CONCLUSIONS: The Proteus syndrome is caused by a somatic activating mutation in AKT1, proving the hypothesis of somatic mosaicism and implicating activation of the PI3K-AKT pathway in the characteristic clinical findings of overgrowth and tumor susceptibility in this disorder. (Funded by the Intramural Research Program of the National Human Genome Research Institute.).


Subject(s)
Mosaicism , Mutation , Proteus Syndrome/genetics , Proto-Oncogene Proteins c-akt/genetics , Child , DNA Mutational Analysis , Exons/genetics , Genotype , Humans , Male , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL