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

Publication year range
1.
Blood ; 139(13): 1939-1953, 2022 03 31.
Article in English | MEDLINE | ID: mdl-35015835

ABSTRACT

Understanding the biological and clinical impact of copy number aberrations (CNAs) on the development of precision therapies in cancer remains an unmet challenge. Genetic amplification of chromosome 1q (chr1q-amp) is a major CNA conferring an adverse prognosis in several types of cancer, including in the blood cancer multiple myeloma (MM). Although several genes across chromosome 1 (chr1q) portend high-risk MM disease, the underpinning molecular etiology remains elusive. Here, with reference to the 3-dimensional (3D) chromatin structure, we integrate multi-omics data sets from patients with MM with genetic variables to obtain an associated clinical risk map across chr1q and to identify 103 adverse prognosis genes in chr1q-amp MM. Prominent among these genes, the transcription factor PBX1 is ectopically expressed by genetic amplification and epigenetic activation of its own preserved 3D regulatory domain. By binding to reprogrammed superenhancers, PBX1 directly regulates critical oncogenic pathways and a FOXM1-dependent transcriptional program. Together, PBX1 and FOXM1 activate a proliferative gene signature that predicts adverse prognosis across multiple types of cancer. Notably, pharmacological disruption of the PBX1-FOXM1 axis with existing agents (thiostrepton) and a novel PBX1 small molecule inhibitor (T417) is selectively toxic against chr1q-amp myeloma and solid tumor cells. Overall, our systems medicine approach successfully identifies CNA-driven oncogenic circuitries, links them to clinical phenotypes, and proposes novel CNA-targeted therapy strategies in MM and other types of cancer.


Subject(s)
Multiple Myeloma , Chromosomes, Human, Pair 1/metabolism , Forkhead Box Protein M1/genetics , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Pre-B-Cell Leukemia Transcription Factor 1/genetics , Prognosis , Systems Analysis , Transcription Factors/genetics
2.
Clin Immunol ; 183: 8-16, 2017 10.
Article in English | MEDLINE | ID: mdl-28645875

ABSTRACT

The ontogeny of the natural, public IgM repertoire remains incompletely explored. Here, high-resolution immunogenetic analysis of B cells from (unrelated) fetal, child, and adult samples, shows that although fetal liver (FL) and bone marrow (FBM) IgM repertoires are equally diversified, FL is the main source of IgM natural immunity during the 2nd trimester. Strikingly, 0.25% of all prenatal clonotypes, comprising 18.7% of the expressed repertoire, are shared with the postnatal samples, consistent with persisting fetal IgM+ B cells being a source of natural IgM repertoire in adult life. Further, the origins of specific stereotypic IgM+ B cell receptors associated with chronic lymphocytic leukemia, can be traced back to fetal B cell lymphopoiesis, suggesting that persisting fetal B cells can be subject to malignant transformation late in life. Overall, these novel data provide unique insights into the ontogeny of physiological and malignant B lymphopoiesis that spans the human lifetime.


Subject(s)
B-Lymphocytes/immunology , Bone Marrow/immunology , Fetus/immunology , Immunoglobulin Heavy Chains/genetics , Immunoglobulin M/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Liver/immunology , Lymphopoiesis/genetics , Receptors, Antigen, B-Cell/genetics , High-Throughput Nucleotide Sequencing , Humans , Immunoglobulin Heavy Chains/immunology , Immunoglobulin M/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Lymphopoiesis/immunology , Receptors, Antigen, B-Cell/immunology , Sequence Analysis, DNA
3.
Blood ; 124(20): 3151-4, 2014 Nov 13.
Article in English | MEDLINE | ID: mdl-25293775

ABSTRACT

A rare point mutation in the core promoter -270GC-rich box of PIGM, a housekeeping gene, disrupts binding of the generic transcription factor (TF) Sp1 and causes inherited glycosylphosphatidylinositol (GPI) deficiency (IGD). We show that whereas PIGM messenger RNA levels and surface GPI expression in IGD B cells are low, GPI expression is near normal in IGD erythroid cells. This divergent phenotype results from differential promoter chromatin accessibility and binding of Sp1. Specifically, whereas PIGM transcription in B cells is dependent on Sp1 binding to the -270GC-rich box and is associated with lower promoter accessibility, in erythroid cells, Sp1 activates PIGM transcription by binding upstream of (but not to) the -270GC-rich box. These findings explain intact PIGM transcription in IGD erythroid cells and the lack of clinically significant intravascular hemolysis in patients with IGD. Furthermore, they provide novel insights into tissue-specific transcriptional control of a housekeeping gene by a generic TF.


Subject(s)
Glycosylphosphatidylinositols/deficiency , Hemoglobinuria, Paroxysmal/genetics , Mannosyltransferases/genetics , Transcriptional Activation , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Erythrocytes/metabolism , Erythrocytes/pathology , Glycosylphosphatidylinositols/genetics , Glycosylphosphatidylinositols/metabolism , Hemoglobinuria, Paroxysmal/metabolism , Hemoglobinuria, Paroxysmal/pathology , Humans , Mutation , Phenotype , Promoter Regions, Genetic , Seizures , Sp1 Transcription Factor/metabolism
4.
J Immunol ; 186(5): 2910-7, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21278350

ABSTRACT

Invariant NKT (iNKT) cells modulate innate and adaptive immune responses through activation of myeloid dendritic cells and macrophages and via enhanced clonogenicity, differentiation, and egress of their shared myeloid progenitors. Because these same progenitors give rise to osteoclasts (OCs), which also mediate the egress of hematopoietic progenitors and orchestrate bone remodeling, we hypothesized that iNKT cells would extend their myeloid cell regulatory role to the development and function of OCs. In this study, we report that selective activation of iNKT cells by α-galactosylceramide causes myeloid cell egress, enhances OC progenitor and precursor development, modifies the intramedullary kinetics of mature OCs, and enhances their resorptive activity. OC progenitor activity is positively regulated by TNF-α and negatively regulated by IFN-γ, but is IL-4 and IL-17 independent. These data demonstrate a novel role of iNKT cells that couples osteoclastogenesis with myeloid cell egress in conditions of immune activation.


Subject(s)
Cell Differentiation/immunology , Lymphocyte Activation/immunology , Natural Killer T-Cells/cytology , Natural Killer T-Cells/immunology , Osteoclasts/immunology , Osteoclasts/metabolism , Animals , Cell Movement/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Down-Regulation/immunology , Interferon-gamma/physiology , Macrophage Colony-Stimulating Factor/physiology , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/cytology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Natural Killer T-Cells/metabolism , Osteoclasts/cytology , RANK Ligand/physiology , Stem Cells/cytology , Stem Cells/immunology , Stem Cells/metabolism , Tumor Necrosis Factor-alpha/physiology , Up-Regulation/immunology
5.
Nat Commun ; 12(1): 5450, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34521827

ABSTRACT

Multiple myeloma is a genetically heterogeneous cancer of the bone marrow plasma cells (PC). Distinct myeloma transcriptome profiles are primarily driven by myeloma initiating events (MIE) and converge into a mutually exclusive overexpression of the CCND1 and CCND2 oncogenes. Here, with reference to their normal counterparts, we find that myeloma PC enhanced chromatin accessibility combined with paired transcriptome profiling can classify MIE-defined genetic subgroups. Across and within different MM genetic subgroups, we ascribe regulation of genes and pathways critical for myeloma biology to unique or shared, developmentally activated or de novo formed candidate enhancers. Such enhancers co-opt recruitment of existing transcription factors, which although not transcriptionally deregulated per se, organise aberrant gene regulatory networks that help identify myeloma cell dependencies with prognostic impact. Finally, we identify and validate the critical super-enhancer that regulates ectopic expression of CCND2 in a subset of patients with MM and in chronic lymphocytic leukemia.


Subject(s)
Carcinogenesis/genetics , Cyclin D1/genetics , Cyclin D2/genetics , Gene Expression Regulation, Neoplastic , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Multiple Myeloma/genetics , Transcriptome , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Carcinogenesis/metabolism , Carcinogenesis/pathology , Case-Control Studies , Cell Line, Tumor , Chromatin/chemistry , Chromatin/metabolism , Cyclin D1/metabolism , Cyclin D2/metabolism , Enhancer Elements, Genetic , Gene Expression Profiling , Gene Regulatory Networks , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Multiple Myeloma/metabolism , Multiple Myeloma/mortality , Multiple Myeloma/pathology , Plasma Cells/metabolism , Plasma Cells/pathology , Proto-Oncogene Proteins c-maf/genetics , Proto-Oncogene Proteins c-maf/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Survival Analysis
6.
Biol Blood Marrow Transplant ; 16(2): 263-72, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19822218

ABSTRACT

We conducted a prospective, multicenter investigation of human-leukocyte antigen (HLA) identical sibling bone marrow transplantation (BMT) in children with severe sickle cell disease (SCD) between 1991 and 2000. To determine if children were protected from complications of SCD after successful BMT, we extended our initial study of BMT for SCD to conduct assessments of the central nervous system (CNS) and of pulmonary function 2 or more years after transplantation. In addition, the impact on gonadal function was studied. After BMT, patients with stroke who had stable engraftment of donor cells experienced no subsequent stroke events after BMT, and brain magnetic resonance imaging (MRI) exams demonstrated stable or improved appearance. However, 2 patients with graft rejection had a second stroke after BMT. After transplantation, most patients also had unchanged or improved pulmonary function. Among the 11 patients who had restrictive lung changes at baseline, 5 were improved and 6 had persistent restrictive disease after BMT. Of the 2 patients who had obstructive changes at baseline, 1 improved and 1 had worsened obstructive disease after BMT. There was, however, significant gonadal toxicity after BMT, particularly among female recipients. In summary, individuals who had stable donor engraftment did not experience sickle-related complications after BMT, and were protected from progressive CNS and pulmonary disease.


Subject(s)
Anemia, Sickle Cell/therapy , Bone Marrow Transplantation/adverse effects , Central Nervous System Diseases/etiology , Gonadal Disorders/etiology , Health Status , Lung Diseases, Obstructive/etiology , Adolescent , Anemia, Sickle Cell/complications , Anemia, Sickle Cell/physiopathology , Central Nervous System Diseases/physiopathology , Child , Donor Selection , Female , Follow-Up Studies , Gonadal Disorders/physiopathology , Graft Survival , Graft vs Host Disease/drug therapy , Graft vs Host Disease/etiology , Graft vs Host Disease/mortality , HLA Antigens/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Histocompatibility Testing , Humans , Lung Diseases, Obstructive/physiopathology , Male , Siblings , Survival Analysis , Transplantation Chimera , Treatment Outcome
7.
N Engl J Med ; 356(16): 1641-7, 2007 Apr 19.
Article in English | MEDLINE | ID: mdl-17442906

ABSTRACT

Disrupted binding of the transcription factor Sp1 to the mutated promoter region of the mannosyl transferase-encoding gene PIGM causes inherited glycosylphosphatidylinositol (GPI) deficiency characterized by splanchnic vein thrombosis and epilepsy. We show that this results in histone hypoacetylation at the promoter of PIGM. The histone deacetylase inhibitor butyrate increases PIGM transcription and surface GPI expression in vitro as well as in vivo through enhanced histone acetylation in an Sp1-dependent manner. More important, the drug caused complete cessation of intractable seizures in a child with inherited GPI deficiency.


Subject(s)
Butyrates/therapeutic use , Glycosylphosphatidylinositols/deficiency , Histone Deacetylase Inhibitors , Mannosyltransferases/genetics , Metabolism, Inborn Errors/drug therapy , Sp1 Transcription Factor/metabolism , Transcription, Genetic/drug effects , Acetylation/drug effects , Adolescent , Butyrates/pharmacology , Epilepsy, Absence/drug therapy , Epilepsy, Absence/etiology , Female , Glycosylphosphatidylinositols/biosynthesis , Humans , Mannosyltransferases/metabolism , Metabolism, Inborn Errors/complications , Metabolism, Inborn Errors/genetics , Mutation , Promoter Regions, Genetic/physiology , Thrombosis/drug therapy , Thrombosis/etiology
8.
J Immunol ; 181(5): 3268-76, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18713998

ABSTRACT

Invariant NKT (iNKT) cells are a subset of highly conserved immunoregulatory T cells that modify a variety of immune responses, including alloreactivity. Central to their function is the interaction of the invariant TCR with glycosphingolipid (GSL) ligands presented by the nonpolymorphic MHC class I molecule CD1d and their ability to secrete rapidly large amounts of immunomodulatory cytokines when activated. Whether iNKT cells, like NK and conventional T cells, can directly display alloreactivity is not known. We show in this study that human iNKT cells and APC can establish a direct cross-talk leading to preferential maturation of allogeneic APC and a considerably higher reactivity of iNKT cells cultured with allogeneic rather that autologous APC. Although the allogeneic activation of iNKT cells is invariant TCR-CD1d interaction-dependent, GSL profiling suggests it does not involve the recognition of disparate CD1d/GSL complexes. Instead, we show that contrary to previous reports, iNKT cells, like NK and T cells, express killer Ig receptors at a frequency similar to that of conventional T cells and that iNKT cell allogeneic activation requires up-regulation and function of activating killer Ig receptors. Thus, iNKT cells can display alloreactivity, for which they use mechanisms characteristic of both NK and conventional T cells.


Subject(s)
Antigens, CD1/metabolism , Isoantigens/immunology , Killer Cells, Natural/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Immunologic/immunology , Antigen Presentation , Antigen-Presenting Cells , Antigens, CD1d , Glycosphingolipids/metabolism , Humans , Lymphocyte Activation , Protein Binding , T-Lymphocytes
9.
Early Hum Dev ; 84(8): 515-23, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18621490

ABSTRACT

The diagnosis, acute management and follow-up of neonates with haemolytic disease of the newborn (HDN) still represents a significant area of activity for maternity/neonatal services. ABO incompatability is now the single largest cause of HDN in the western world. However, with increasing knowledge at the molecular level, and closer liaison between neonatal paediatricians and haematologists, the diagnosis of non-immune causes of HDN is increasing. As these conditions have an inherited basis and therefore have implications for other family members (or future children), it remains a high priority for all neonatal paediatricians to achieve an accurate diagnosis in all cases of HDN. As the efficacy of phototherapy increases the role of exchange transfusion in acute management is rapidly decreasing. This makes gauging the appropriate time to intervene with exchange transfusion a difficult clinical decision, and guidelines appropriate to the spectrum of contemporary disease are required. In the future intravenous immunoglobulin and/or intramuscular metalloporphyrins may further reduce the need for exchange transfusion and continue to change the spectrum of HDN faced by neonatal paediatricians.


Subject(s)
Erythroblastosis, Fetal/therapy , Erythroblastosis, Fetal/blood , Erythroblastosis, Fetal/immunology , Exchange Transfusion, Whole Blood/methods , Exchange Transfusion, Whole Blood/trends , Female , Humans , Immunoglobulins, Intravenous/therapeutic use , Infant, Newborn/immunology , Phototherapy/methods , Phototherapy/standards , Phototherapy/trends , Pregnancy/immunology
10.
Curr Hematol Rep ; 5(1): 55-63, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16537047

ABSTRACT

Neonatal thrombocytopenia is a common clinical problem. The majority of episodes are early-onset thrombocytopenias due to impaired fetal megakaryocytopoiesis associated with placental insufficiency; the commonest causes of severe early-onset thrombocytopenia are immune thrombocytopenias, congenital infections, and asphyxia. By contrast, about 90% of cases of severe thrombocytopenia presenting after the first few days of life are due to late-onset bacterial sepsis, necrotizing enterocolitis, or both. Although clinically stable neonates tolerate relatively low platelet counts without significant risk of hemorrhage, ill or clinically unstable neonates with profound thrombocytopenia often have a poor outcome. Currently, the only therapy is platelet transfusion. Despite many published guidelines for platelet transfusion in the newborn, however, there have been no randomized trials to define the safe lower limit for platelet counts in sick neonates. The platelet threshold at which the benefits of transfusion outweigh the risks in neonates remains unclear. Well-designed trials are urgently needed.


Subject(s)
Thrombocytopenia/etiology , Age of Onset , Blood Transfusion , Humans , Infant, Newborn , Platelet Count , Thrombocytopenia/diagnosis , Thrombocytopenia/therapy
11.
PLoS Med ; 2(2): e44, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15737000

ABSTRACT

BACKGROUND TO THE DEBATE: Umbilical cord blood--the blood that remains in the placenta after birth--can be collected and stored frozen for years. A well-accepted use of cord blood is as an alternative to bone marrow as a source of hematopoietic stem cells for allogeneic transplantation to siblings or to unrelated recipients; women can donate cord blood for unrelated recipients to public banks. However, private banks are now open that offer expectant parents the option to pay a fee for the chance to store cord blood for possible future use by that same child (autologous transplantation).


Subject(s)
Blood Banks/economics , Ethics, Medical , Fetal Blood , Private Sector , Cord Blood Stem Cell Transplantation/ethnology , Humans , Specimen Handling , Transplantation, Autologous
12.
Lancet ; 364(9429): 179-82, 2004.
Article in English | MEDLINE | ID: mdl-15246731

ABSTRACT

Fetal cells enter maternal blood during pregnancy and persist in women with autoimmune disease. The frequency of subsequent fetomaternal microchimerism in healthy women and its cell type is unknown. To test the hypothesis that fetal mesenchymal stem cells persist in maternal organs, we studied female bone marrow and ribs. Male cells were identified by XY fluorescence in-situ hybridisation in marrow-derived mesenchymal stem cells and in rib sections from all women with male pregnancies, but not in controls (9/9 vs 0/5, p=0.0005). We conclude that fetal stem cells transferred into maternal blood engraft in marrow, where they remain throughout life. This finding has implications for normal pregnancy, for obstetric complications that increase fetomaternal trafficking, and for graft survival after transplantation.


Subject(s)
Bone Marrow Cells/cytology , Chimera , Fetus/cytology , Maternal-Fetal Exchange , Mesoderm/cytology , Stem Cells/cytology , Aged , Aged, 80 and over , Bone and Bones/cytology , Chromosomes, Human, X , Chromosomes, Human, Y , Female , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Pregnancy , Ribs , Time Factors
14.
Blood ; 107(8): 3138-44, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16373666

ABSTRACT

Invariant natural killer T cells (iNKT cells) are a small subset of immunoregulatory T cells highly conserved in humans and mice. On activation by glycolipids presented by the MHC-like molecule CD1d, iNKT cells promptly secrete T helper 1 and 2 (Th1/2) cytokines but also cytokines with hematopoietic potential such as GM-CSF. Here, we show that the myeloid clonogenic potential of human hematopoietic progenitors is increased in the presence of glycolipid-activated, GM-CSF-secreting NKT cells; conversely, short- and long-term progenitor activity is decreased in the absence of NKT cells, implying regulation of hematopoiesis in both the presence and the absence of immune activation. In accordance with these findings, iNKT-cell-deficient mice display impaired hematopoiesis characterized by peripheral-blood cytopenias, reduced marrow cellularity, lower frequency of hematopoietic stem cells (HSCs), and reduced early and late hematopoietic progenitors. We also show that CD1d is expressed on human HSCs. CD1d-expressing HSCs display short- and long-term clonogenic potential and can present the glycolipid alpha-galactosylceramide to iNKT cells. Thus, iNKT cells emerge as the first subset of regulatory T cells that are required for effective hematopoiesis in both steady-state conditions and under conditions of immune activation.


Subject(s)
Antigen Presentation/immunology , Galactosylceramides/immunology , Hematopoiesis/immunology , Hematopoietic Stem Cells/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation/physiology , Animals , Antigen Presentation/drug effects , Female , Galactosylceramides/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Hematopoiesis/drug effects , Hematopoietic Stem Cells/cytology , Humans , Killer Cells, Natural/cytology , Lymphocyte Activation/drug effects , Mice , Mice, Knockout , Th1 Cells/cytology , Th1 Cells/immunology , Th2 Cells/cytology , Th2 Cells/immunology
15.
J Immunol ; 175(8): 5087-94, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16210612

ABSTRACT

NKT cells are a small subset of regulatory T cells conserved in humans and mice. In humans they express the Valpha24Jalpha18 invariant chain (hence invariant NKT (iNKT) cells) and are restricted by the glycolipid-presenting molecule CD1d. In mice, iNKT cells may enhance or inhibit anti-infectious and antitumor T cell responses but suppress autoimmune and alloreactive responses. We postulated that iNKT cells might also modulate human alloreactive responses. Using MLR assays we demonstrate that in the presence of the CD1d-presented glycolipid alpha-galactosylceramide (alphaGC) alloreactivity is enhanced (37 +/- 12%; p < 0.001) in an iNKT cell-dependent manner. iNKT cells are activated early during the course of the MLR, presumably by natural ligands. In MLR performed without exogenous ligands, depletion of iNKT cells significantly diminished the alloresponse in terms of proliferation (58.8 +/- 24%; p < 0.001) and IFN-gamma secretion (43.2 +/- 15.2%; p < 0.001). Importantly, adding back fresh iNKT cells restored the reactivity of iNKT cell-depleted MLR to near baseline levels. CD1d-blocking mAbs equally reduced the reactivity of the iNKT cell-replete and -depleted MLR compared with IgG control, indicating that the effect of iNKT cells in the in vitro alloresponse is CD1d-dependent. These findings suggest that human iNKT cells, although not essential for its development, can enhance the alloreactive response.


Subject(s)
Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Antigens, CD1/physiology , Antigens, CD1d , CD3 Complex/metabolism , Cell Proliferation , Cells, Cultured , Flow Cytometry , Galactosylceramides/physiology , Humans , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Ligands , Lymphocyte Culture Test, Mixed , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
16.
Curr Opin Pediatr ; 15(1): 17-23, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12544267

ABSTRACT

Thrombocytopenia remains a common problem in sick newborns. A quarter of all neonates admitted to neonatal intensive care units develop thrombocytopenia, and in 20% of episodes the thrombocytopenia is severe (platelets <50 x 10(9)/L). Practical and clinically relevant classifications of neonatal thrombocytopenia have now been developed which, by highlighting the principal conditions precipitating severe thrombocytopenia (eg, sepsis, necrotizing enterocolitis, perinatal asphyxia, and the immune thrombocytopenias), aid the practicing neonatologist. Recent reviews demonstrate that many neonates with severe thrombocytopenia receive repeated platelet transfusions, although evidence of their clinical benefit is lacking, and there exists a significant variation in platelet transfusion practice between centers. These facts support the need for the development of evidence-based protocols for platelet transfusion in the newborn and stimulate continued interest in the potential of hemopoietic growth factors (, thrombopoietin and interleukin-11) to prevent or treat neonatal thrombocytopenia.


Subject(s)
Thrombocytopenia/diagnosis , Thrombocytopenia/therapy , Age Factors , Humans , Infant, Newborn , Risk Factors , Thrombocytopenia/etiology
17.
Am J Obstet Gynecol ; 188(4): 1002-10, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12712101

ABSTRACT

OBJECTIVE: The purpose of this study was to investigate whether purified CD34(+) cells from first-trimester fetal blood are a source of primitive and committed hemopoietic progenitors. STUDY DESIGN: CD34(+) cells from first-trimester fetal blood and term cord blood were assayed for committed hemopoietic progenitor cells, high proliferative potential colony-forming cells, and long-term culture-initiating cells. RESULTS: First-trimester CD34(+) cells that were compared with cells at term generated fewer hemopoietic progenitor cells and fewer high proliferative potential colony-forming cells with lower recloning efficiency(P <.001). First-trimester CD34(+) cells tended to contain more long-term culture-initiating cells, both in bulk cultures and by limiting dilution analysis. The ratio between committed and primitive progenitors was 3 in the first-trimester and 20 in the term cord blood, respectively. CONCLUSION: First-trimester fetal blood is enriched in primitive (compared with committed) hemopoietic progenitors and may be an advantageous source of stem cells for prenatal therapy.


Subject(s)
Antigens, CD34/analysis , Blood Cells/cytology , Blood Cells/immunology , Fetal Blood , Hematopoietic Stem Cells/cytology , Cells, Cultured , Colony-Forming Units Assay , Female , Humans , Pregnancy , Pregnancy Trimester, First , Time Factors
18.
Pediatr Res ; 51(6): 756-60, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12032273

ABSTRACT

Thrombocytopenia is common in sick neonates, and affected neonates have adverse outcomes compared with those without thrombocytopenia. As impaired platelet production underlies many neonatal thrombocytopenias, affected neonates are potential candidates for hemopoietic growth factor therapy. Although recombinant human (rh) thrombopoietin remains under therapeutic development, rhIL-11, which stimulates megakaryocytopoiesis and increases platelet counts after chemotherapy, is already licensed for clinical use. However, nothing is known about IL-11 in neonates. We therefore measured plasma IL-11 by ELISA in healthy term neonates, stable preterm neonates with or without thrombocytopenia, and preterm neonates with sepsis or necrotizing enterocolitis (NEC) with or without thrombocytopenia. At birth IL-11 was undetectable (<10 pg/mL) in healthy term neonates (n = 20) and 27 of 31 (87%) stable preterm neonates. Three stable preterm neonates with detectable IL-11 (mean+/-SD, 11.3 +/- 0.4 pg/mL; median, 11.6 pg/mL) suffered chorioamnionitis, the remaining neonate (IL-11, 14 pg/mL) being one of nine with early onset thrombocytopenia (present by <72 h of age). IL-11 was also measured in 58 preterm neonates with suspected sepsis or NEC. In 25 of 58, sepsis or NEC was unconfirmed and IL-11 was undetectable. By contrast, 14 of 33 with proven sepsis or NEC had elevated IL-11 (median, 14.9 pg/mL; range, 11.2-92.2 pg/mL). Of these 33 neonates, 19 developed thrombocytopenia: nine of 19 (47%) had detectable IL-11 and 10 of 19 (53%) did not (p > 0.05). Although its role in platelet production in neonates remains unclear, these data suggest that IL-11 is involved in the endogenous cytokine response to sepsis or NEC in preterm neonates. Further studies of IL-11 in neonates are warranted to assess its role both in platelet production and in mediation of the endogenous inflammatory response.


Subject(s)
Infant, Newborn, Diseases/blood , Infant, Newborn, Diseases/immunology , Interleukin-11/blood , Thrombocytopenia/blood , Thrombocytopenia/immunology , Enterocolitis, Necrotizing/blood , Enterocolitis, Necrotizing/immunology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Infant, Newborn , Male , Sepsis/blood , Sepsis/immunology
19.
Br J Haematol ; 120(2): 289-95, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12542489

ABSTRACT

Stem cell transplantation (SCT) remains the only cure for thalassaemia major. Recent advances in medical treatment make it even more important that accurate information is available regarding outcome of SCT in relevant patient populations in order to guide informed decisions regarding the most appropriate treatment for individual thalassaemia patients. We report the results of 55 consecutive first related allogeneic bone marrow transplants (BMT) for children with beta-thalassaemia major performed in two UK paediatric centres over 10 years. Between February 1991 and February 2001, 55 children underwent 57 allogeneic BMT. The median age at BMT was 6.4 years and the majority of patients (73%) originated from the Indian subcontinent. Using the Pesaro risk classification, 17 patients were class 1, 27 were class 2 and 11 were class 3. Actuarial overall survival and thalassaemia-free survival at 8 years were 94.5% (95% CI 85.1-98.1) and 81.8% (95% CI 69.7-89.8) respectively. Despite the majority of patients being in class 2 or 3, transplant-related mortality was low (5.4%). The principal complication was graft rejection accompanied by autologous reconstitution that occurred in 13.2% of transplants. Following modification of the conditioning regimen in 1993, the rejection rate fell to 4.6% and remained low. Acute graft-versus-host disease (GVHD) of grade II-IV occurred in 31% and chronic GVHD in 14.5%. These data compare favourably with survival with medical treatment for thalassaemia major and suggest that allogeneic BMT remains an important treatment option for children with beta-thalassaemia major, particularly when compliance with iron chelation is poor.


Subject(s)
Bone Marrow Transplantation/methods , beta-Thalassemia/therapy , Acute Disease , Child , Chronic Disease , Disease-Free Survival , Female , Graft Rejection , Graft vs Host Disease , Humans , Male , Survival Rate , Transplantation, Homologous , beta-Thalassemia/mortality
20.
Prenat Diagn ; 22(6): 463-9, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12116304

ABSTRACT

OBJECTIVES: Selective amplification of rare fetal cells in maternal blood is a potential strategy for non-invasive prenatal diagnosis. We assessed the proliferative potential of first trimester fetal progenitors compared to maternal ones. METHODS: Fetal and maternal haemopoietic progenitors were cultured separately and in two model mixtures: (i) co-cultures of male fetal nucleated cells mixed with maternal nucleated cells and (ii) co-cultures of malefetal CD34+ cells with maternal CD34+ cells. Cell origin was detected by X-Y fluorescence in situ hybridisation (FISH) RESULTS: The frequency of haemopoietic progenitors in first trimester fetal blood (predominantly CFU-GEMM) differed from those in peripheral blood from pregnant women (predominantly BFU-e). First trimester haemopoietic progenitors formed larger colonies (p=0.0001) and their haemoglobinisation was accelerated compared to those of maternal origin (p<0.001). CD34+ fetal haemopoietic progenitor cells could be expanded four times more than their maternal counterparts (median 235.8-fold, range 174.0-968.0 vs 71.9-fold, range 41.1-192.0; p=0.003). While selective expansion of fetal cells was not observed in the mononuclear cell model, the CD34+ cell rare event mixtures produced a 463.2-fold (range 128.0-2915.0) expansion of fetal cells. CONCLUSION: Selective expansion of first trimester fetal haemopoietic progenitors may be useful for amplifying fetal cells from maternal blood.


Subject(s)
Cell Division , Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Prenatal Diagnosis/methods , Antigens, CD34/analysis , Cell Count , Cells, Cultured , Coculture Techniques , Colony-Forming Units Assay , Erythroid Precursor Cells/cytology , Female , Hematopoietic Stem Cells/immunology , Hemoglobins/metabolism , Humans , In Situ Hybridization, Fluorescence , Pregnancy , Pregnancy Trimester, First
SELECTION OF CITATIONS
SEARCH DETAIL