Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
1.
Ann Hematol ; 103(6): 1887-1896, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38581547

ABSTRACT

We evaluated the impact of the genotype on clinical and hematochemical features, hepatic and cardiac iron levels, and endocrine, hepatic, and cardiovascular complications in non-transfusion-dependent (NTD) ß-thalassemia intermedia (TI) patients. Sixty patients (39.09 ± 11.11 years, 29 females) consecutively enrolled in the Myocardial Iron Overload in Thalassemia project underwent Magnetic Resonance Imaging to quantify iron overload, biventricular function parameters, and atrial areas and to detect replacement myocardial fibrosis. Three groups of patients were identified: homozygous ß+ (N = 18), heterozygous ß0ß+ (N = 22), and homozygous ß0 (N = 20). The groups were homogeneous for sex, age, splenectomy, hematochemical parameters, chelation therapy, and iron levels. The homozygous ß° genotype was associated with significantly higher biventricular end-diastolic and end-systolic volume indexes and bi-atrial area indexes. No difference was detected in biventricular ejection fractions or myocardial fibrosis. Extramedullary hematopoiesis and leg ulcers were significantly more frequent in the homozygous ß° group compared to the homozygous ß+ group. No association was detected between genotype and liver cirrhosis, hypogonadism, hypothyroidism, osteoporosis, heart failure, arrhythmias, and pulmonary hypertension. Heart remodelling related to a high cardiac output state cardiomyopathy, extramedullary hematopoiesis, and leg ulcers were more pronounced in patients with the homozygous ß° genotype compared to the other genotypes analyzed. The knowledge of the genotype can assist in the clinical management of NTD ß-TI patients.


Subject(s)
Genotype , Iron Overload , Iron , beta-Thalassemia , Humans , beta-Thalassemia/genetics , beta-Thalassemia/complications , Female , Male , Adult , Middle Aged , Iron Overload/genetics , Iron Overload/etiology , Iron/metabolism , Leg Ulcer/etiology , Leg Ulcer/genetics , Hematopoiesis, Extramedullary/genetics , Magnetic Resonance Imaging , Myocardium/pathology , Myocardium/metabolism , Liver Cirrhosis/genetics , Liver Cirrhosis/complications , Homozygote
2.
EMBO Rep ; 24(12): e57485, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-37870318

ABSTRACT

Bacterial infections can activate and mobilize hematopoietic stem and progenitor cells (HSPCs) from the bone marrow (BM) to the spleen, a process termed extramedullary hematopoiesis (EMH). Recent studies suggest that commensal bacteria regulate not only the host immune system but also hematopoietic homeostasis. However, the impact of gut microbes on hematopoietic pathology remains unclear. Here, we find that systemic single injections of Akkermansia muciniphila (A. m.), a mucin-degrading bacterium, rapidly activate BM myelopoiesis and slow but long-lasting hepato-splenomegaly, characterized by the expansion and differentiation of functional HSPCs, which we term delayed EMH. Mechanistically, delayed EMH triggered by A. m. is mediated entirely by the MYD88/TRIF innate immune signaling pathway, which persistently stimulates splenic myeloid cells to secrete interleukin (IL)-1α, and in turn, activates IL-1 receptor (IL-1R)-expressing splenic HSPCs. Genetic deletion of Toll-like receptor-2 and -4 (TLR2/4) or IL-1α partially diminishes A. m.-induced delayed EMH, while inhibition of both pathways alleviates splenomegaly and EMH. Our results demonstrate that cooperative IL-1R- and TLR-mediated signals regulate commensal bacteria-driven EMH, which might be relevant for certain autoimmune disorders.


Subject(s)
Hematopoiesis, Extramedullary , Humans , Hematopoiesis, Extramedullary/genetics , Splenomegaly/metabolism , Bone Marrow , Hematopoietic Stem Cells/metabolism , Hematopoiesis
3.
Tohoku J Exp Med ; 256(2): 119-125, 2022 02.
Article in English | MEDLINE | ID: mdl-35173090

ABSTRACT

Although splenomegaly is one of the important signs of primary myelofibrosis, the differential diagnosis varies from malignant disorders to benign disorders, including malignant lymphoma and sarcoidosis. The patient was a 67-year-old male who developed anemia and huge splenomegaly. The laboratory findings include human T-cell leukemia virus type 1 (HTLV-1) antibody, elevated soluble interleukin-2 receptor, hypocellular bone marrow, and uptake in the spleen on positron emission tomography/computed tomography scan. Additionally, we performed laparoscopic splenectomy to alleviate the clinical symptoms and to rule out malignant lymphoma. Histological findings revealed extramedullary hematopoiesis, characterized by the presence of erythroid islands and clusters of dysplastic megakaryocytes with increased reticulin fibrosis. Immunohistochemical staining revealed the presence of von Willebrand factor, dysplastic megakaryocytes, myeloperoxidase, myeloid-predominant proliferations, and CD34 immature myeloid cells. Furthermore, regarding the angiogenesis in the spleen, the endothelial cells of the capillaries and those of the sinusoidal vascular system that were reactive for CD34 and CD8, respectively, were also detected. Consequently, the histological findings revealed both extramedullary hematopoiesis and angiogenesis in spleen. Based on the histological findings and the identification of Janus activating kinase 2 (JAK-2) mutation, the patient was diagnosed with primary myelofibrosis. Splenectomy reduces blood transfusion requirements after surgery. The patient was carefully followed-up without further treatments. Thus, primary myelofibrosis is the crucial differential diagnosis of huge splenomegaly.


Subject(s)
Hematopoiesis, Extramedullary , Primary Myelofibrosis , Aged , Endothelial Cells , Hematopoiesis, Extramedullary/genetics , Humans , Male , Primary Myelofibrosis/genetics , Primary Myelofibrosis/pathology , Spleen/pathology , Splenomegaly/pathology
4.
Int J Hematol ; 115(6): 898-901, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35089531

ABSTRACT

We present the case of a 56-year-old male patient with paravertebral extramedullary hematopoiesis (EMH) secondary to myelodysplastic syndrome with ring sideroblasts and multilineage dysplasia. In a routine health checkup over 5 years prior, he presented with asymptomatic mild anemia and a posterior mediastinal mass. Pathological and cytomorphological findings of the resected paravertebral mass were similar to those of his bone marrow specimen, and included cellularity with erythroid hyperplasia, multilineage dysplastic changes, and the presence of ring sideroblasts. A concordant SF3B1 mutation was detected in both bone marrow and paravertebral mass samples, suggesting that the EMH cells were derived from the bone marrow.


Subject(s)
Hematopoiesis, Extramedullary , Myelodysplastic Syndromes , Hematopoiesis, Extramedullary/genetics , Humans , Male , Middle Aged , Mutation , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Phosphoproteins/genetics , RNA Splicing Factors/genetics
5.
Med Mol Morphol ; 55(1): 68-75, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34396490

ABSTRACT

Myelofibrosis is characterized by stem cell-derived clonal proliferation potentially resulting in bone marrow fibrosis. As the disease progresses, extramedullary hematopoiesis is frequently detected in the spleen and the liver but rarely in other organs. We report a case of a 68-year-old woman with myelofibrosis with a JAK2 mutation, showing extramedullary hematopoiesis (EMH) in various organs with a marked increase in reticulin fibers, and myeloproliferative neoplasm (MPN)-related necrotizing crescent glomerulonephritis. She was admitted to our hospital owing to respiratory discomfort. Computed tomography revealed a mass in the anterior mediastinum. Ten days later, the patient died owing to respiratory distress. At autopsy, EMH were detected in the anterior mediastinum, heart, lung, spleen, and the kidney with a marked increase in reticulin fibers. We considered that respiratory distress was partially caused by EMH. In the kidney, necrotizing crescent glomerulonephritis was observed. Immunohistochemically, the glomerular basement and mesangial area were IgA- and C3d-positive. Ultrastructural examination revealed the presence of dense deposits in the subendothelial space and the mesangial and paramesangial areas. Thus, we suspected that MPN-related necrotizing crescentic glomerulonephritis harbored a pathogenesis similar to that of IgA-dominant post-infectious glomerulonephritis or IgA nephropathy. This case report could widen the spectrum of MPN- or EMH-related lesions.


Subject(s)
Hematopoiesis, Extramedullary , Primary Myelofibrosis , Aged , Autopsy , Female , Hematopoiesis, Extramedullary/genetics , Hemorrhage , Humans , Primary Myelofibrosis/diagnosis , Primary Myelofibrosis/genetics , Primary Myelofibrosis/pathology , Reticulin
6.
Cells ; 10(6)2021 06 15.
Article in English | MEDLINE | ID: mdl-34203913

ABSTRACT

Glomerular kidney disease causing nephrotic syndrome is a complex systemic disorder and is associated with significant morbidity in affected patient populations. Despite its clinical relevance, well-established models are largely missing to further elucidate the implications of uncontrolled urinary protein loss. To overcome this limitation, we generated a novel, inducible, podocyte-specific transgenic mouse model (Epb41l5fl/fl*Nphs1-rtTA-3G*tetOCre), developing nephrotic syndrome in adult mice. Animals were comprehensively characterized, including microbiome analysis and multiplexed immunofluorescence imaging. Induced knockout mice developed a phenotype consistent with focal segmental glomerular sclerosis (FSGS). Although these mice showed hallmark features of severe nephrotic syndrome (including proteinuria, hypoalbuminemia and dyslipidemia), they did not exhibit overt chronic kidney disease (CKD) phenotypes. Analysis of the gut microbiome demonstrated distinct dysbiosis and highly significant enrichment of the Alistipes genus. Moreover, Epb41l5-deficient mice developed marked organ pathologies, including extramedullary hematopoiesis of the spleen. Multiplex immunofluorescence imaging demonstrated red pulp macrophage proliferation and mTOR activation as driving factors of hematopoietic niche expansion. Thus, this novel mouse model for adult-onset nephrotic syndrome reveals the significant impact of proteinuria on extra-renal manifestations, demonstrating the versatility of this model for nephrotic syndrome-related research.


Subject(s)
Hematopoiesis, Extramedullary/physiology , Nephrotic Syndrome/metabolism , Nephrotic Syndrome/microbiology , Animals , Disease Models, Animal , Dysbiosis/pathology , Gastrointestinal Microbiome/genetics , Gastrointestinal Microbiome/physiology , Glomerulosclerosis, Focal Segmental/pathology , Hematopoiesis, Extramedullary/genetics , Humans , Kidney Diseases/physiopathology , Macrophages/metabolism , Mice , Mice, 129 Strain , Mice, Knockout , Mice, Transgenic , Models, Biological , Nephrotic Syndrome/physiopathology , Podocytes/metabolism , Primary Cell Culture , Proteins/metabolism , Proteinuria/metabolism
7.
Virchows Arch ; 479(4): 755-764, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33934231

ABSTRACT

BCR-ABL-fusion-negative myeloproliferative neoplasms (MPNs) with myelofibrosis (MF) include primary MF, post-polycythemia vera MF and post-essential thrombocythemia MF. Clonal extramedullary hematopoiesis (EMH) can occur during MPN pathogenesis. Although histopathological bone-marrow (BM) features during clonal EMH have been investigated, those of the spleen have been poorly described. We analyzed splenectomy samples from 28 patients with MF and BM samples from 20 of them. Slides were stained with hematoxylin and eosin, reticulin, and trichrome, with immunohistochemical labeling of glycophorin A, myeloperoxidase, CD61, CD34, and CD117. We also subjected splenectomy and BM samples from six patients and spleen samples from seven patients to next-generation sequencing (NGS). Megakaryocyte-rich spleen nodules (MRSNs), seen in seven of the 28 patients, were significantly associated with megakaryocyte proliferation in the spleen (p = 0.04). We devised a grading system for spleen fibrosis (SF) and found that SF was increased in 20 of 28 patients. Notably, patients with SF were more likely to have MRSNs, suggesting that megakaryocytes might participate in SF, as previously described in BM. Comparisons of spleen and BM NGS findings of six patients' specimens revealed identical mutational status in the two organs for half of the patients. We observed additional mutations in the spleen of two patients. However, the meaning of this finding remains unknown since there was a long interval between BM and spleen samplings (68 and 82 months, respectively).


Subject(s)
Hematopoiesis, Extramedullary/physiology , Primary Myelofibrosis/pathology , Adult , Aged , Aged, 80 and over , Bone Marrow/pathology , Disease Progression , Female , France/epidemiology , Hematopoiesis , Hematopoiesis, Extramedullary/genetics , Humans , Male , Middle Aged , Myeloproliferative Disorders/epidemiology , Myeloproliferative Disorders/pathology , Polycythemia Vera/pathology , Primary Myelofibrosis/epidemiology , Primary Myelofibrosis/genetics , Spleen/pathology , Thrombocythemia, Essential/pathology
8.
Semin Diagn Pathol ; 37(6): 296-302, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32591154

ABSTRACT

Myeloid diseases detected as primary or secondary lesions in the lung and pleura are rare. Clinical presentations and radiographic results may vary significantly depending on the nature of the diseases. The most common diseases associated with lung and pleura involvement are myeloid sarcoma/acute myeloid leukemia (AML) and extramedullary hematopoiesis (EMH). AML typically represents localized involvement by systemic acute leukemia, while EMH is frequently secondary to underlying benign hematolymphoid disorders or myeloproliferative neoplasms. This review provides an overview of the pathogenesis, clinical presentations, radiologic/imaging studies, pathologic and genetic findings, and treatment/outcomes associated with myeloid diseases in the lung and pleura.


Subject(s)
Lung/pathology , Myeloproliferative Disorders , Pleura/pathology , Diagnosis, Differential , Genetic Predisposition to Disease , Hematopoiesis, Extramedullary/genetics , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/therapy , Myeloproliferative Disorders/diagnosis , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Myeloproliferative Disorders/therapy , Pathology, Molecular , Sarcoma, Myeloid/diagnosis , Sarcoma, Myeloid/genetics , Sarcoma, Myeloid/pathology , Sarcoma, Myeloid/therapy , Thalassemia/diagnosis , Thalassemia/genetics , Thalassemia/pathology , Thalassemia/therapy , Treatment Outcome
10.
Cell Mol Life Sci ; 77(14): 2723-2738, 2020 Jul.
Article in English | MEDLINE | ID: mdl-31974657

ABSTRACT

Extramedullary hematopoiesis (EMH) is the expansion and differentiation of hematopoietic stem and progenitor cells outside of the bone marrow. In postnatal life, as a compensatory mechanism for ineffective hematopoiesis of the bone marrow, pathological EMH is triggered by hematopoietic disorders, insufficient hematopoietic compensation, and other pathological stress conditions, such as infection, advanced tumors, anemia, and metabolic stress. Pathological EMH has been reported in many organs, and the sites of pathological EMH may be related to reactivation of the embryonic hematopoietic structure in these organs. As a double-edged sword (blood and immune cell supplementation as well as clinical complications), pathological EMH has been widely studied in recent years. In particular, pathological EMH induced by late-stage tumors contributes to tumor immunosuppression. Thus, a deeper understanding of the mechanism of pathological EMH may be conducive to the development of therapies against the pathological processes that induce EMH. This article reviews the recent progress of research on the cellular and molecular mechanisms of pathological EMH in specific diseases.


Subject(s)
Embryonic Stem Cells , Hematopoiesis, Extramedullary/genetics , Hematopoietic Stem Cells , Neoplasms/genetics , Humans , Immunosuppression Therapy , Neoplasms/pathology , Stress, Physiological/genetics
11.
Biomed Res Int ; 2019: 4504302, 2019.
Article in English | MEDLINE | ID: mdl-30834265

ABSTRACT

BACKGROUND: Extramedullary hematopoiesis (EMH) is common in non-transfusion-dependent thalassemia (NTDT) patients. Clinical presentations of EMH vary as MRI screening is not feasible. Hence, serum biomarkers are used to predict the risk of EMH. MATERIALS AND METHODS: 52 NTDT patients, including 26 EMH (+) and 26 EMH (-), together with 26 healthy controls, were enrolled in this case-control study from 2013 to 2016. EMH was confirmed by computed tomography or MRI. Demographic, transfusion, genetic, laboratory, and liver iron concentration (LIC) data, as well as clinical complications, were analyzed. RESULTS: EMH (+) patients had significantly higher serum ferritin (SF), growth differentiation factor 15 (GDF15), and erythropoietin (EPO) levels compared with EMH (-) patients and controls. The levels of erythroferrone (ERFE), hepcidin, and sTfR did not differ significantly between EMH (+) and EMH (-) patients (p>0.05). In NTDT patients, serum ERFE was not related to SF, LIC, hepcidin, sTfR, EPO, GDF15, and Hb levels. GDF15, EPO concentrations, and GDF15 to sTfR and GDF15 to EPO ratios are able to determine the presence of EMH with considerable sensitivity and specificity. CONCLUSIONS: GDF15, EPO, and GDF15 to EPO and GDF15 to sTfR ratios are potential biomarkers for the early prediction of NTDT in patients who are at risk for EMH.


Subject(s)
Antigens, CD/blood , Erythropoietin/blood , Growth Differentiation Factor 15/blood , Hematopoiesis, Extramedullary/genetics , Receptors, Transferrin/blood , Thalassemia/blood , Adult , Biomarkers/blood , Case-Control Studies , Erythropoiesis/genetics , Female , Ferritins/blood , Hematopoiesis, Extramedullary/physiology , Hepcidins/blood , Homeostasis/genetics , Humans , Iron/metabolism , Magnetic Resonance Imaging , Male , Middle Aged , Peptide Hormones/blood , Risk Factors , Thalassemia/complications , Thalassemia/diagnostic imaging , Thalassemia/physiopathology , Tomography, X-Ray Computed
14.
Blood ; 131(20): 2223-2234, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29555646

ABSTRACT

Despite the well-established cell-intrinsic role of epigenetic factors in normal and malignant hematopoiesis, their cell-extrinsic role remains largely unexplored. Herein we investigated the hematopoietic impact of inactivating Ezh2, a key component of polycomb repressive complex 2 (PRC2), in the fetal liver (FL) vascular niche. Hematopoietic specific (Vav-iCre) Ezh2 inactivation enhanced FL hematopoietic stem cell (HSC) expansion with normal FL erythropoiesis. In contrast, endothelium (Tie2-Cre) targeted Ezh2 inactivation resulted in embryonic lethality with severe anemia at embryonic day 13.5 despite normal emergence of functional HSCs. Ezh2-deficient FL endothelium overexpressed Mmp9, which cell-extrinsically depleted the membrane-bound form of Kit ligand (mKitL), an essential hematopoietic cytokine, in FL. Furthermore, Mmp9 inhibition in vitro restored mKitL expression along with the erythropoiesis supporting capacity of FL endothelial cells. These data establish that Ezh2 is intrinsically dispensable for FL HSCs and provides proof of principle that modulation of epigenetic regulators in niche components can exert a marked cell-extrinsic impact.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Fetus , Hematopoiesis, Extramedullary , Liver/physiology , Anemia/genetics , Anemia/metabolism , Animals , Biomarkers , Cells, Cultured , Enhancer of Zeste Homolog 2 Protein/metabolism , Fluorescent Antibody Technique , Gene Expression , Gene Silencing , Hematopoiesis, Extramedullary/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Immunohistochemistry , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Phenotype , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Stem Cell Factor/metabolism
16.
J Thromb Haemost ; 15(11): 2226-2229, 2017 11.
Article in English | MEDLINE | ID: mdl-28921865

ABSTRACT

Essentials Extramedullary hematopoiesis (EMH) represents a pathologic finding in adult life. We report a mass-like EMH in the presacral space in a patient with ANKRD26-related thrombocytopenia. We found possible correlation between EMH and conditions causing lifelong thrombocytopenia. EMH can cause masses of unknown origin in patients with inherited thrombocytopenias. SUMMARY: Most commonly located in the liver and spleen, extramedullary hematopoiesis (EMH) is the presence of hematopoietic tissue outside the bone marrow. MYH9-related thrombocytopenia (MYH9-RD) and ANKRD26-related thrombocytopenia (ANKRD26-RT) are two of the most frequent forms of inherited thrombocytopenia (IT). Until recently, EMH has been associated with neoplastic and non-neoplastic hematologic conditions in which ITs were not included. We describe a case of mass-like EMH in the presacral space in a patient affected with ANKRD26-RT, comparing it with another case of paravertebral EMH we recently described in a subject with MYH9-RD. The surprisingly similitude of such a finding in the context of a group of rare disorders induces us to speculate about the possible pathogenic relationship between EMH and conditions causing lifelong thrombocytopenia, particularly the entity of ITs. Finally, we suggest that EMH has to be taken into consideration in the diagnostic work-up of masses of unknown origin in subjects affected with ITs.


Subject(s)
Hematopoiesis, Extramedullary/genetics , Mutation , Nuclear Proteins/genetics , Thrombocytopenia/genetics , Aged , Biopsy, Needle , Bone Marrow Examination , DNA Mutational Analysis , Female , Genetic Predisposition to Disease , Heredity , Humans , Immunohistochemistry , Intercellular Signaling Peptides and Proteins , Pelvis , Phenotype , Positron Emission Tomography Computed Tomography , Spleen/diagnostic imaging , Thrombocytopenia/blood , Thrombocytopenia/diagnosis
17.
J Exp Med ; 214(10): 2875-2887, 2017 Oct 02.
Article in English | MEDLINE | ID: mdl-28878000

ABSTRACT

Recent advances in single-cell, transcriptomic profiling have provided unprecedented access to investigate cell heterogeneity during tissue and organ development. In this study, we used massively parallel, single-cell RNA sequencing to define cell heterogeneity within the zebrafish kidney marrow, constructing a comprehensive molecular atlas of definitive hematopoiesis and functionally distinct renal cells found in adult zebrafish. Because our method analyzed blood and kidney cells in an unbiased manner, our approach was useful in characterizing immune-cell deficiencies within DNA-protein kinase catalytic subunit (prkdc), interleukin-2 receptor γ a (il2rga), and double-homozygous-mutant fish, identifying blood cell losses in T, B, and natural killer cells within specific genetic mutants. Our analysis also uncovered novel cell types, including two classes of natural killer immune cells, classically defined and erythroid-primed hematopoietic stem and progenitor cells, mucin-secreting kidney cells, and kidney stem/progenitor cells. In total, our work provides the first, comprehensive, single-cell, transcriptomic analysis of kidney and marrow cells in the adult zebrafish.


Subject(s)
Hematopoiesis, Extramedullary/genetics , Kidney/cytology , RNA/genetics , Zebrafish/anatomy & histology , Animals , Animals, Genetically Modified , Cell Lineage/genetics , Cell Lineage/physiology , Gene Expression Profiling , Hematopoiesis, Extramedullary/physiology , Hematopoietic Stem Cells , Kidney/metabolism , Sequence Analysis, RNA , Zebrafish/genetics , Zebrafish/metabolism
18.
PLoS One ; 12(7): e0181881, 2017.
Article in English | MEDLINE | ID: mdl-28732075

ABSTRACT

The purpose of this study was to perform a comprehensive evaluation and selection of reference genes for the study of extramedullary hematopoiesis during development and the early post-natal period. A total of six candidate reference genes (ACTB, GAPDH, HPRT1, PPID, TBP, TUBB3) in four organs (heart, liver, spleen, and thymus) over five perinatal time points (Embryonic days 14.5, 16.5, 18.5, Post-natal days 0, 21) were evaluated by quantitative real-time PCR. The expression stability of the candidate reference genes were analyzed using geNorm, NormFinder, Bestkeeper, Delta CT method, and RefFinder software packages. Detailed methodology for isolation of high quality/purity RNA and analysis is presented. Detailed analysis demonstrated that TBP is the best single reference gene for embryonic samples and HPRT1 is the best single reference gene for post-natal and pooled embryonic and post-natal samples. Organ-level analysis demonstrated that HPRT1 was the most suitable reference gene for heart, liver and thymus samples, while TBP was the best candidate for spleen samples. In general, TUBB3 was consistently the least stable gene for normalization. This is the first study to describe a systematic comprehensive selection of reference genes for murine extramedullary hematopoietic tissues over a developmental time course. We provide suggested reference genes for individual tissues and developmental stages and propose that a combination of reference genes affords flexibility in experimental design and analysis.


Subject(s)
Gene Expression/genetics , Hematopoiesis, Extramedullary/genetics , Animals , Gene Expression Profiling/methods , Mice , Mice, Inbred C57BL , RNA/genetics , Real-Time Polymerase Chain Reaction/methods , Reference Standards
20.
Acta Haematol ; 137(3): 175-182, 2017.
Article in English | MEDLINE | ID: mdl-28399542

ABSTRACT

Extramedullary hematopoiesis (EMH) results from the extension of hematopoietic tissue beyond the confines of the bones. Since the initiation of regular transfusion programs from an early age for all thalassemia major (ΤΜ) patients, EMH has not been considered a clinical issue anymore. The present study aims to record the prevalence of EMH in chronically transfused ΤΜ patients followed at our institution and to investigate possible risk factors associated with its occurrence. The project was designed as a retrospective, nonexperimental, descriptive, exploratory study. In total, the study enrolled 104 patients. EMH was revealed in 15/104 (14%) patients. The presence of intravening sequence (IVS)-I-6 was significantly related with the development of EMH (p < 0.05). No other demographic or biological factor studied was found to be related with the presence of EMH. The study stresses a profound incidence of asymptomatic EMH in a solid group of well-transfused ΤΜ patients. Given the high incidence of the IVS-I-6 allele in the Mediterranean and Middle Eastern region, high-quality, prospective, multicenter studies could confirm the association of EMH occurrence with the presence of the IVS-I-6 mutation and further evaluate the exact role of this mutation in the EMH process.


Subject(s)
Hematopoiesis, Extramedullary/genetics , Mutation , beta-Globins/genetics , beta-Thalassemia/genetics , beta-Thalassemia/pathology , Adult , Alleles , Female , Genotype , Greece/epidemiology , Humans , Incidence , Male , Middle Aged , Retrospective Studies , Risk Factors , Young Adult , beta-Thalassemia/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...