Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 35
1.
Leuk Res ; 105: 106570, 2021 06.
Article En | MEDLINE | ID: mdl-33838549

Transient abnormal myelopoiesis (TAM) in neonates with Down syndrome, which spontaneously resolves within several weeks or months after birth, may represent a special form of leukemia developing in the fetal liver (FL). To explore the role of hepatoblasts, one of the major constituents of the FL hematopoietic microenvironment, in the pathogenesis of TAM, we investigated the influence of a human hepatoblastoma cell line, HUH-6, on the in vitro growth and differentiation of TAM blasts. In a coculture system with membrane filters, which hinders cell-to-cell contact between TAM blasts and HUH-6 cells, the growth and megakaryocytic differentiation of TAM blast progenitors were increased in the presence of HUH-6 cells. The culture supernatant of HUH-6 cells contained hematopoietic growth factors, including stem cell factor (SCF) and thrombopoietin (TPO). The neutralizing antibody against SCF abrogated the growth-stimulating activity of the culture supernatant of HUH-6 cells, demonstrating that, among the growth factors produced by HUH-6 cells, SCF may be the major growth stimulator and that TPO may be involved in megakaryocytic differentiation, rather than growth, of TAM blasts. This suggests that hepatoblasts function in the regulation of the growth and differentiation of TAM blasts in the FL through the production of hematopoietic growth factors, including SCF and TPO, and are involved in the leukemogenesis of TAM.


Down Syndrome/pathology , Hepatoblastoma , Hepatocytes , Leukemoid Reaction/pathology , Megakaryocyte Progenitor Cells , Cell Differentiation/physiology , Cell Line, Tumor , Cell Proliferation/physiology , Coculture Techniques , Hepatoblastoma/metabolism , Hepatoblastoma/pathology , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Leukemia, Megakaryoblastic, Acute/pathology , Megakaryocyte Progenitor Cells/metabolism , Megakaryocyte Progenitor Cells/pathology , Myelopoiesis/physiology , Stem Cells/metabolism , Stem Cells/pathology
3.
Int J Hematol ; 110(1): 119-123, 2019 Jul.
Article En | MEDLINE | ID: mdl-30879266

A 42-year-old female complaining of fever and night sweats was diagnosed with acute megakaryoblastic blast phase chronic myeloid leukemia (CML-BP). She had massive splenomegaly, left pleural effusion with leukemia infiltration, and moderate myelofibrosis. She received dasatinib monotherapy (140 mg/day) as for induction, after which her pleural effusion rapidly resolved and hematological remission was achieved. However, CML relapsed 4 months after starting dasatinib due to increased BCR-ABL fusion signals in the peripheral blood. The T315I mutation was also detected at the recurrence of CML. As a salvage treatment, ponatinib monotherapy (45 mg/day) was started immediately. After 5 months, BCR-ABL fusion signals decreased to 5%, and myelofibrosis improved from MF Grade 2 to 1; she then underwent allogeneic bone marrow transplantation from an unrelated donor. However, the graft failed, and cord blood transplantation (CBT) was performed. Ponatinib (15 mg/day) was continued after CBT as a maintenance treatment, with molecular complete response continuing for more than 1 year with no severe adverse events, including cardiovascular events. There is limited evidence regarding the optimal dose and schedule of ponatinib before and after allogeneic hematopoietic stem cell transplantation, especially in patients with CML-BP having T315I mutation; thus, well-designed clinical trials are warranted.


Hematopoietic Stem Cell Transplantation/methods , Imidazoles/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Mutation , Pyridazines/therapeutic use , Adult , Blast Crisis/pathology , Cord Blood Stem Cell Transplantation/methods , Female , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Megakaryocyte Progenitor Cells/pathology , Recurrence , Salvage Therapy/methods , Transplantation, Homologous , Treatment Outcome
6.
Hematol Oncol Stem Cell Ther ; 11(3): 178-182, 2018 Sep.
Article En | MEDLINE | ID: mdl-29684340

Myeloid sarcoma is an extramedullary tumor that consists of myeloblasts or immature myeloid cells. The neoplasm can occur in any part of the body, including the bone, periosteum, lymph nodes, skin, and soft tissue and they may occur de novo or in association with acute myeloid leukemia, myeloproliferative neoplasms and myelodysplastic syndromes. Most cases display a myelomonocytic or pure monoblastic morphology. Tumors with megakaryoblastic differentiation are extremely uncommon and may occur in association with transformation of a myeloproliferative disorder. Myeloid sarcoma presenting as a breast mass is very rare and diagnostically challenging. We report a case of myeloid sarcoma with megakaryoblastic differentiation in the breast of a patient with history of essential thrombocythemia. The mass was composed of undifferentiated pleomorphic malignant cells in trabecular cords and nests with many scattered giant malignant cells and brisk abnormal mitoses. On immunohistochemistry, the neoplastic cells were positive for CD61, CD31, CD34, Factor VIII and CD43 which confirmed the diagnosis. To our knowledge, this is the first report of myeloid sarcoma with megakaryoblastic morphology occurring in the breast. Here we discuss the clinicopathologic features of this rare entity and the challenges involved in making this difficult diagnosis.


Antigens, Differentiation/metabolism , Breast Neoplasms , Cell Differentiation , Megakaryocyte Progenitor Cells , Neoplasm Proteins/metabolism , Sarcoma, Myeloid , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Humans , Megakaryocyte Progenitor Cells/metabolism , Megakaryocyte Progenitor Cells/pathology , Middle Aged , Sarcoma, Myeloid/metabolism , Sarcoma, Myeloid/pathology
7.
J Pharmacol Sci ; 135(1): 14-21, 2017 Sep.
Article En | MEDLINE | ID: mdl-28927780

Metformin has received increasing attention for its potential anticancer activity against certain human leukemia cells, but its effects on human megakaryoblastic cells are unclear. This study aimed to investigate the effects of metformin on proliferation and apoptosis of human megakaryoblastic cells (Dami and MEG-01) and the underlying molecular mechanisms. CCK8 assay was employed to measure cell proliferation. Flow cytometry was adopted to detect cell apoptosis. Western blot was further employed to measure apoptosis-related proteins. In Dami and MEG-01 cells, metformin significantly inhibited proliferation and promoted apoptosis in a dose- and time-dependent manner, and metformin (4 mM) was selected for subsequent experiments. Metformin inhibited ERK1/2, JNK, and PI3K/Akt, but activated p38 pathway in these two cells. Moreover, inhibition of ERK1/2, JNK or PI3K/Akt pathway alone induced cell apoptosis compared to the control group. The combination of specific inhibitors of ERK1/2, JNK or PI3K/Akt pathway and metformin further promoted cell apoptosis and the up-regulation of p21, Bax, Bad, cleaved caspase-3 and -9 as well as the down-regulation of Bcl-2 mediated by metformin alone, but inhibition of p38 pathway exhibited the opposite results. These findings support the possibility of metformin treatment as a new therapeutic strategy against acute megakaryoblastic leukemia (AMKL).


Apoptosis/drug effects , Apoptosis/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Hypoglycemic Agents/pharmacology , Megakaryocyte Progenitor Cells/cytology , Megakaryocyte Progenitor Cells/pathology , Metformin/pharmacology , Cell Line , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Hypoglycemic Agents/therapeutic use , JNK Mitogen-Activated Protein Kinases , Leukemia, Megakaryoblastic, Acute/drug therapy , Leukemia, Megakaryoblastic, Acute/genetics , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/physiology , Metformin/therapeutic use , Molecular Targeted Therapy , Phosphatidylinositol 3-Kinases , Signal Transduction/drug effects , Signal Transduction/genetics , p38 Mitogen-Activated Protein Kinases
9.
Blood ; 130(3): 271-284, 2017 07 20.
Article En | MEDLINE | ID: mdl-28490570

RUNX1 is crucial for the regulation of megakaryocyte specification, maturation, and thrombopoiesis. Runx1 possesses 2 promoters: the distal P1 and proximal P2 promoters. The major protein isoforms generated by P1 and P2 are RUNX1C and RUNX1B, respectively, which differ solely in their N-terminal amino acid sequences. RUNX1C is the most abundantly expressed isoform in adult hematopoiesis, present in all RUNX1-expressing populations, including the cKit+ hematopoietic stem and progenitor cells. RUNX1B expression is more restricted, being highly expressed in the megakaryocyte lineage but downregulated during erythropoiesis. We generated a Runx1 P1 knock-in of RUNX1B, termed P1-MRIPV This mouse line lacks RUNX1C expression but has normal total RUNX1 levels, solely comprising RUNX1B. Using this mouse line, we establish a specific requirement for the P1-RUNX1C isoform in megakaryopoiesis, which cannot be entirely compensated for by RUNX1B overexpression. P1 knock-in megakaryocyte progenitors have reduced proliferative capacity and undergo increased cell death, resulting in thrombocytopenia. P1 knock-in premegakaryocyte/erythroid progenitors demonstrate an erythroid-specification bias, evident from increased erythroid colony-forming ability and decreased megakaryocyte output. At a transcriptional level, multiple erythroid-specific genes are upregulated and megakaryocyte-specific transcripts are downregulated. In addition, proapoptotic pathways are activated in P1 knock-in premegakaryocyte/erythroid progenitors, presumably accounting for the increased cell death in the megakaryocyte progenitor compartment. Unlike in the conditional adult Runx1 null models, megakaryocytic maturation is not affected in the P1 knock-in mice, suggesting that RUNX1B can regulate endomitosis and thrombopoiesis. Therefore, despite the high degree of structural similarity, RUNX1B and RUNX1C isoforms have distinct and specific roles in adult megakaryopoiesis.


Apoptosis Regulatory Proteins/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Megakaryocyte Progenitor Cells/metabolism , Megakaryocytes/metabolism , RNA, Messenger/genetics , Thrombocytopenia/genetics , Thrombopoiesis/genetics , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins/metabolism , Cell Death , Cell Lineage/genetics , Cell Proliferation , Core Binding Factor Alpha 2 Subunit/metabolism , Flow Cytometry , Gene Expression Regulation , Humans , Immunophenotyping , Megakaryocyte Progenitor Cells/pathology , Megakaryocytes/pathology , Mice , Mice, Transgenic , Promoter Regions, Genetic , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Thrombocytopenia/metabolism , Thrombocytopenia/pathology
10.
Blood ; 129(25): 3332-3343, 2017 06 22.
Article En | MEDLINE | ID: mdl-28336526

The developmental pathway for human megakaryocytes remains unclear, and the definition of pure unipotent megakaryocyte progenitor is still controversial. Using single-cell transcriptome analysis, we have identified a cluster of cells within immature hematopoietic stem- and progenitor-cell populations that specifically expresses genes related to the megakaryocyte lineage. We used CD41 as a positive marker to identify these cells within the CD34+CD38+IL-3RαdimCD45RA- common myeloid progenitor (CMP) population. These cells lacked erythroid and granulocyte-macrophage potential but exhibited robust differentiation into the megakaryocyte lineage at a high frequency, both in vivo and in vitro. The efficiency and expansion potential of these cells exceeded those of conventional bipotent megakaryocyte/erythrocyte progenitors. Accordingly, the CD41+ CMP was defined as a unipotent megakaryocyte progenitor (MegP) that is likely to represent the major pathway for human megakaryopoiesis, independent of canonical megakaryocyte-erythroid lineage bifurcation. In the bone marrow of patients with essential thrombocythemia, the MegP population was significantly expanded in the context of a high burden of Janus kinase 2 mutations. Thus, the prospectively isolatable and functionally homogeneous human MegP will be useful for the elucidation of the mechanisms underlying normal and malignant human hematopoiesis.


Hematopoiesis , Megakaryocyte Progenitor Cells/cytology , Megakaryocyte Progenitor Cells/metabolism , Megakaryocytes/cytology , Adult , Animals , Antigens, CD/analysis , Cell Lineage , Cells, Cultured , Humans , Megakaryocyte Progenitor Cells/pathology , Megakaryocytes/metabolism , Mice, Inbred C57BL , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Platelet Membrane Glycoprotein IIb/analysis , Transcriptome
11.
Blood ; 127(26): 3410-23, 2016 06 30.
Article En | MEDLINE | ID: mdl-27081096

An activating JAK2V617F mutation has been found in ∼50% patients with myelofibrosis (MF). Inactivating mutations in histone methyltransferase enhancer of zeste homolog 2 (EZH2) also have been observed in patients with MF. Interestingly, inactivating EZH2 mutations are often associated with JAK2V617F mutation in MF, although their contributions in the pathogenesis of MF remain elusive. To determine the effects of concomitant loss of EZH2 and JAK2V617F mutation in hematopoiesis, we generated Ezh2-deficient Jak2V617F-expressing mice. Whereas expression of Jak2V617F alone induced a polycythemia vera-like disease, concomitant loss of Ezh2 significantly reduced the red blood cell and hematocrit parameters but increased the platelet counts in Jak2V617F knock-in mice. Flow cytometric analysis showed impairment of erythroid differentiation and expansion of megakaryocytic precursors in Ezh2-deficient Jak2V617F mice. Moreover, loss of Ezh2 enhanced the repopulation capacity of Jak2V617F-expressing hematopoietic stem cells. Histopathologic analysis revealed extensive fibrosis in the bone marrow (BM) and spleen of Ezh2-deleted Jak2V617F mice. Transplantation of BM from Ezh2-deleted Jak2V617F mice into wild-type animals resulted in even faster progression to MF. Gene expression profiling and chromatin immunoprecipitation sequence analysis revealed that S100a8, S100a9, Ifi27l2a, and Hmga2 were transcriptionally derepressed, and the H3K27me3 levels in these gene promoters were significantly reduced on Ezh2 deletion in hematopoietic progenitors of Jak2V617F mice. Furthermore, overexpression of S100a8, S100a9, Ifi27l2a, or Hmga2 significantly increased megakaryocytic colonies in the BM of Jak2V617F mice, indicating a role for these Ezh2 target genes in altered megakaryopoiesis involved in MF. Overall, our results suggest that loss of Ezh2 cooperates with Jak2V617F in the development of MF in Jak2V617F-expressing mice.


Enhancer of Zeste Homolog 2 Protein/deficiency , Hematologic Neoplasms , Janus Kinase 2/metabolism , Mutation, Missense , Primary Myelofibrosis/metabolism , Amino Acid Substitution , Animals , Gene Deletion , Gene Expression Regulation, Neoplastic , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Janus Kinase 2/genetics , Megakaryocyte Progenitor Cells/metabolism , Megakaryocyte Progenitor Cells/pathology , Mice , Mice, Transgenic , Platelet Count , Primary Myelofibrosis/genetics , Primary Myelofibrosis/pathology
12.
Blood ; 127(10): 1260-8, 2016 Mar 10.
Article En | MEDLINE | ID: mdl-26787735

Hematopoietic stem cells (HSCs) can be safely collected from the body, genetically modified, and re-infused into a patient with the goal to express the transgene product for an individual's lifetime. Hematologic defects that can be corrected with an allogeneic bone marrow transplant can theoretically also be treated with gene replacement therapy. Because some genetic disorders affect distinct cell lineages, researchers are utilizing HSC gene transfer techniques using lineage-specific endogenous gene promoters to confine transgene expression to individual cell types (eg, ITGA2B for inherited platelet defects). HSCs appear to be an ideal target for platelet gene therapy because they can differentiate into megakaryocytes which are capable of forming several thousand anucleate platelets that circulate within blood vessels to establish hemostasis by repairing vascular injury. Platelets play an essential role in other biological processes (immune response, angiogenesis) as well as diseased states (atherosclerosis, cancer, thrombosis). Thus, recent advances in genetic manipulation of megakaryocytes could lead to new and improved therapies for treating a variety of disorders. In summary, genetic manipulation of megakaryocytes has progressed to the point where clinically relevant strategies are being developed for human trials for genetic disorders affecting platelets. Nevertheless, challenges still need to be overcome to perfect this field; therefore, strategies to increase the safety and benefit of megakaryocyte gene therapy will be discussed.


Genetic Therapy/methods , Hematologic Diseases/therapy , Hematopoietic Stem Cell Transplantation , Megakaryocyte Progenitor Cells/metabolism , Megakaryocytes/metabolism , Animals , Hematologic Diseases/genetics , Hematologic Diseases/metabolism , Hematologic Diseases/pathology , Humans , Integrin alpha2/biosynthesis , Integrin alpha2/genetics , Megakaryocyte Progenitor Cells/pathology , Megakaryocytes/pathology , Transgenes
13.
Hematol Oncol Stem Cell Ther ; 9(1): 30-3, 2016 Mar.
Article En | MEDLINE | ID: mdl-26169238

Basophilia is commonly associated with chronic myelogenous leukemia, notably in the accelerated phase or during blast crisis. It is also associated with other myeloproliferative neoplasms. However, its association with acute leukemia is very rare and is described in association with acute basophilic leukemia and few acute myeloid leukemias (AMLs) with recurrent genetic abnormalities such as t(6;9)(p23;q34). Herein, we describe the morphological features and discuss the differential diagnosis of a case of AML with the blasts showing previously unreported unusual combination of megakaryoblastic and basophilic differentiation along with peripheral blood and bone marrow basophilia.


Basophils/pathology , Bone Marrow/pathology , Leukemia, Myeloid, Acute/pathology , Megakaryocyte Progenitor Cells/pathology , Adolescent , Diagnosis, Differential , Female , Humans , Leukemia, Myeloid, Acute/diagnosis
14.
Cell Stem Cell ; 17(4): 422-34, 2015 Oct 01.
Article En | MEDLINE | ID: mdl-26299573

Infections are associated with extensive platelet consumption, representing a high risk for health. However, the mechanism coordinating the rapid regeneration of the platelet pool during such stress conditions remains unclear. Here, we report that the phenotypic hematopoietic stem cell (HSC) compartment contains stem-like megakaryocyte-committed progenitors (SL-MkPs), a cell population that shares many features with multipotent HSCs and serves as a lineage-restricted emergency pool for inflammatory insults. During homeostasis, SL-MkPs are maintained in a primed but quiescent state, thus contributing little to steady-state megakaryopoiesis. Even though lineage-specific megakaryocyte transcripts are expressed, protein synthesis is suppressed. In response to acute inflammation, SL-MkPs become activated, resulting in megakaryocyte protein production from pre-existing transcripts and a maturation of SL-MkPs and other megakaryocyte progenitors. This results in an efficient replenishment of platelets that are lost during inflammatory insult. Thus, our study reveals an emergency machinery that counteracts life-threatening platelet depletions during acute inflammation.


Blood Platelets/pathology , Inflammation/pathology , Megakaryocyte Progenitor Cells/pathology , Thrombopoiesis , Animals , Blood Platelets/physiology , Cell Lineage , Cell Proliferation , Hematopoietic Stem Cells/pathology , Hematopoietic Stem Cells/physiology , Megakaryocyte Progenitor Cells/physiology , Mice
15.
Bioorg Med Chem ; 23(10): 2387-407, 2015 May 15.
Article En | MEDLINE | ID: mdl-25882525
16.
Eur J Haematol ; 94(5): 456-62, 2015 May.
Article En | MEDLINE | ID: mdl-24853125

Transient myeloproliferative disorder (TMD) is a clonal proliferation of megakaryoblasts, typically occurring in newborns with Down syndrome. It is believed that TMD occurs in the presence of GATA1 mutation together with trisomy 21. However, a limited number of patients with TMD but without Down syndrome have been reported, all with a blast population with numeric or rarely structural chromosome 21 abnormalities. We present the first case of a newborn boy with a TMD without trisomy 21 and without any of the mentioned molecular or cytogenetic abnormalities. This case report suggests that unknown disease mechanisms may provoke or mimic TMD. This case report is followed by a concise review of the literature discussing the different entities and pathomechanisms of TMD and acute megakaryocytic leukaemia in patients with or without Down syndrome.


Down Syndrome/diagnosis , Down Syndrome/pathology , Leukemoid Reaction/diagnosis , Leukemoid Reaction/pathology , Megakaryocyte Progenitor Cells/pathology , Down Syndrome/genetics , Down Syndrome/therapy , Humans , Infant, Newborn , Leukemoid Reaction/genetics , Leukemoid Reaction/therapy , Male , Megakaryocyte Progenitor Cells/metabolism , Platelet Transfusion
17.
Arch Pathol Lab Med ; 138(10): 1302-6, 2014 Oct.
Article En | MEDLINE | ID: mdl-25268193

Transient abnormal myelopoiesis occurs exclusively in patients with Down syndrome (constitutional trisomy 21), manifests in the neonatal period, and is characterized by circulating megakaryoblasts with varied degrees of multisystem organ involvement. In most cases, this process resolves spontaneously by 3 to 6 months of age, but for some, the disease can be fatal. Affected patients are particularly prone to develop acute megakaryoblastic leukemia in early childhood. Somatic GATA1 mutations are believed to be pivotal in the development of transient abnormal myelopoiesis and have proven to be a marker of clonal identity in its evolution to megakaryoblastic leukemia. We describe a study case of transient abnormal myelopoiesis and review the clinical manifestations, laboratory features, natural history, molecular genetics, and postulated disease pathogenesis of this disorder.


Biomarkers, Tumor/genetics , Down Syndrome/diagnosis , GATA1 Transcription Factor/genetics , Leukemia, Megakaryoblastic, Acute/diagnosis , Megakaryocyte Progenitor Cells/pathology , Mutation , Neoplasm Proteins/genetics , Biomarkers, Tumor/blood , Down Syndrome/blood , Down Syndrome/genetics , Down Syndrome/pathology , GATA1 Transcription Factor/blood , Humans , Infant, Newborn , Leukemia, Megakaryoblastic, Acute/blood , Leukemia, Megakaryoblastic, Acute/genetics , Leukemia, Megakaryoblastic, Acute/pathology , Male , Megakaryocyte Progenitor Cells/metabolism , Neoplasm Proteins/blood
20.
Hematology ; 19(7): 417-23, 2014 Oct.
Article En | MEDLINE | ID: mdl-24295040

OBJECTIVES: Thrombocytopenia is common in HIV-infected individuals and often requires a diagnostic bone marrow examination. Interpretation may, however, be limited due to the multifactorial nature of HIV-associated thrombocytopenia and the difficulty in assessing megakaryocyte function morphologically. The immature platelet fraction (IPF) is a parameter which reportedly reflects megakaryocyte activity, with an IPF >7.7% suggesting increased platelet production. The aim of this study was to correlate the IPF with the bone marrow findings as well as other clinical variables of interest in South African patients with HIV-associated thrombocytopenia. METHODS: Seventy-eight HIV-positive patients with thrombocytopenia were enrolled from the Charlotte Maxeke Johannesburg Academic Hospital. The IPF levels were measured using a Sysmex XE-5000 haematology analyzer and were correlated with bone marrow and other findings. RESULTS: The median IPF was 7.6%, ranging from 1.3 to 44%. It was raised in 78% of patients with immune thrombocytopenia (ITP) (median = 16.3%) and low in 79% of patients with hypocellular marrow (median = 6.5%). Surprisingly, it was highly variable among patients with malignant marrow infiltration and mycobacterial infection of the bone marrow (BMTB) (median = 8.4 and 7.1%, respectively). Multivariate linear regression analysis confirmed a significant independent inverse relationship between the IPF and hypocellular marrow (P < 0.0001), a marginally significant positive association with ITP (P = 0.059), and the absence of any relationship with malignant infiltration or BMTB. The IPF had a significant inverse association with the platelet count (P = 0.0006), but was unrelated to the CD4 count and exposure to anti-retroviral therapy. Unexpectedly, it showed a significant positive association with the HIV viral load (P = 0.005). We speculate this to reflect increased megakaryocyte activity in compensation for accelerated platelet clearance due to HIV-driven platelet activation. CONCLUSION: This study investigates the role of the IPF in HIV-associated thrombocytopenia, and emphasizes the limitations of morphological analysis in determining megakaryocyte function.


Bone Marrow Diseases/blood , HIV Infections/blood , Megakaryocyte Progenitor Cells/pathology , Thrombocytopenia/blood , Adult , Bone Marrow Diseases/virology , Cell Count , Female , HIV Infections/virology , Humans , Linear Models , Male , Megakaryocytes/pathology , Multivariate Analysis , Platelet Count , South Africa , Thrombocytopenia/virology , Viral Load
...