Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 280
Filter
1.
PLoS Biol ; 21(5): e3001746, 2023 05.
Article in English | MEDLINE | ID: mdl-37134077

ABSTRACT

Extramedullary hematopoiesis (EMH) expands hematopoietic capacity outside of the bone marrow in response to inflammatory conditions, including infections and cancer. Because of its inducible nature, EMH offers a unique opportunity to study the interaction between hematopoietic stem and progenitor cells (HSPCs) and their niche. In cancer patients, the spleen frequently serves as an EMH organ and provides myeloid cells that may worsen pathology. Here, we examined the relationship between HSPCs and their splenic niche in EMH in a mouse breast cancer model. We identify tumor produced IL-1α and leukemia inhibitory factor (LIF) acting on splenic HSPCs and splenic niche cells, respectively. IL-1α induced TNFα expression in splenic HSPCs, which then activated splenic niche activity, while LIF induced proliferation of splenic niche cells. IL-1α and LIF display cooperative effects in activating EMH and are both up-regulated in some human cancers. Together, these data expand avenues for developing niche-directed therapies and further exploring EMH accompanying inflammatory pathologies like cancer.


Subject(s)
Hematologic Diseases , Hematopoiesis, Extramedullary , Neoplasms , Humans , Animals , Mice , Hematopoiesis, Extramedullary/physiology , Leukemia Inhibitory Factor/pharmacology , Interleukin-1alpha/pharmacology , Hematopoiesis
2.
Blood ; 136(1): 119-129, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32202634

ABSTRACT

Abnormal megakaryocyte development and platelet production lead to thrombocytopenia or thrombocythemia and increase the risk of hemorrhage or thrombosis. Acylglycerol kinase (AGK) is a mitochondrial membrane kinase that catalyzes the formation of phosphatidic acid and lysophosphatidic acid. Mutation of AGK has been described as the major cause of Sengers syndrome, and the patients with Sengers syndrome have been reported to exhibit thrombocytopenia. In this study, we found that megakaryocyte/platelet-specific AGK-deficient mice developed thrombocytopenia and splenomegaly, mainly caused by inefficient bone marrow thrombocytopoiesis and excessive extramedullary hematopoiesis, but not by apoptosis of circulating platelets. It has been reported that the G126E mutation arrests the kinase activity of AGK. The AGK G126E mutation did not affect peripheral platelet counts or megakaryocyte differentiation, suggesting that the involvement of AGK in megakaryocyte development and platelet biogenesis was not dependent on its kinase activity. The Mpl/Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (Stat3) pathway is the major signaling pathway regulating megakaryocyte development. Our study confirmed that AGK can bind to JAK2 in megakaryocytes/platelets. More interestingly, we found that the JAK2 V617F mutation dramatically enhanced the binding of AGK to JAK2 and greatly facilitated JAK2/Stat3 signaling in megakaryocytes/platelets in response to thrombopoietin. We also found that the JAK2 JAK homology 2 domain peptide YGVCF617CGDENI enhanced the binding of AGK to JAK2 and that cell-permeable peptides containing YGVCF617CGDENI sequences accelerated proplatelet formation. Therefore, our study reveals critical roles of AGK in megakaryocyte differentiation and platelet biogenesis and suggests that targeting the interaction between AGK and JAK2 may be a novel strategy for the treatment of thrombocytopenia or thrombocythemia.


Subject(s)
Mutation, Missense , Phosphotransferases (Alcohol Group Acceptor)/physiology , Point Mutation , Splenomegaly/genetics , Thrombocytopenia/genetics , Thrombopoiesis/physiology , Amino Acid Sequence , Animals , Blood Platelets/enzymology , Cells, Cultured , Hematopoiesis, Extramedullary/physiology , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Liver/cytology , Liver/embryology , Megakaryocytes/enzymology , Mice , Mice, Knockout , Mitochondrial Membranes/enzymology , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Binding , Protein Interaction Mapping , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Splenomegaly/enzymology , Thrombocytopenia/enzymology , Thrombopoiesis/drug effects
3.
Pharmacol Res ; 169: 105663, 2021 07.
Article in English | MEDLINE | ID: mdl-33979688

ABSTRACT

Recruitment of innate immune cells and their accumulation in the arterial wall and infarcted myocardium has been recognized as a central feature of atherosclerosis and cardiac ischemic injury, respectively. In both, steady state and under pathological conditions, majority of these cells have a finite life span and are continuously replenished from haematopoietic stem/progenitor cell pool residing in the bone marrow and extramedullary sites. While having a crucial role in the cardiovascular disease development, proliferation and differentiation of innate immune cells within haematopoietic compartments is greatly affected by the ongoing cardiovascular pathology. In the current review, we summarize key cells, processes and tissue compartments that are involved in myelopoiesis under the steady state, during atherosclerosis development and in myocardial infarction.


Subject(s)
Bone Marrow/physiopathology , Cardiovascular Diseases/physiopathology , Hematopoiesis, Extramedullary/physiology , Myelopoiesis/physiology , Animals , Atherosclerosis/physiopathology , Humans , Myeloid Cells/physiology
4.
Clin Radiol ; 75(8): 565-578, 2020 08.
Article in English | MEDLINE | ID: mdl-31973940

ABSTRACT

Extramedullary haematopoiesis (EMH) refers to the formation of non-neoplastic blood cell lines outside the bone marrow and is a common incidental finding when patients with haematological disorders are imaged. EMH presenting as mass (tumefactive EMH) has long been a radiological conundrum as it resembles neoplasms. Several imaging findings have been described in EMH, and these vary depending on the activity of the underlying haematopoiesis. The older lesions are easier to diagnose as they often demonstrate characteristic findings such as haemosiderin and fat deposition. In comparison, the newer, actively haematopoietic lesions often mimic neoplasms. Molecular imaging, particularly 99mTc labelled sulphur colloid scintigraphy, may be helpful in such cases. Although imaging is extremely useful in detecting and characterising EMH, imaging alone is often non-diagnostic as no single mass shows all the typical findings. Hence, a judgement based on the clinical background, combination of imaging findings, and slow interval growth may be more appropriate and practical in making the correct diagnosis. In every case, an effort has to be made in providing an imaging-based diagnosis as it may prevent a potentially risky biopsy. When confident differentiation is not possible, biopsy has to be resorted to. This article describes the causes, pathophysiology, and theories underlying the genesis of EMH, followed by the general and location-specific imaging findings. The purpose is to provide a thorough understanding of the condition as well as enable the clinical radiologist in making an imaging-based diagnosis whenever possible and identify the situations where biopsy has to be performed.


Subject(s)
Bone Marrow/diagnostic imaging , Hematologic Diseases/diagnosis , Hematopoiesis, Extramedullary/physiology , Tomography, X-Ray Computed/methods , Biopsy , Humans
5.
Dev Biol ; 434(2): 292-303, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29253505

ABSTRACT

The embryonic aorta produces hematopoietic stem and progenitor cells from a hemogenic endothelium localized in the aortic floor through an endothelial to hematopoietic transition. It has been long proposed that the Bone Morphogenetic Protein (BMP)/Transforming Growth Factor ß (TGFß) signaling pathway was implicated in aortic hematopoiesis but the very nature of the signal was unknown. Here, using thorough expression analysis of the BMP/TGFß signaling pathway members in the endothelial and hematopoietic compartments of the aorta at pre-hematopoietic and hematopoietic stages, we show that the TGFß pathway is preferentially balanced with a prominent role of Alk1/TgfßR2/Smad1 and 5 on both chicken and mouse species. Functional analysis using embryonic stem cells mutated for Acvrl1 revealed an enhanced propensity to produce hematopoietic cells. Collectively, we reveal that TGFß through the Alk1/TgfßR2 receptor axis is acting on endothelial cells to produce hematopoiesis.


Subject(s)
Aorta/embryology , Avian Proteins/metabolism , Endothelium, Vascular/embryology , Hematopoiesis, Extramedullary/physiology , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Animals , Aorta/cytology , Chick Embryo , Chickens , Endothelium, Vascular/cytology , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism , Smad1 Protein/metabolism , Smad5 Protein/metabolism
6.
J Pak Med Assoc ; 69(7): 1059, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31983750

ABSTRACT

Hodgkin lymphoma is a high grade lymphoma which is usually confined to the lymphnodes. Extranodal involvement of the Hodgkin lymphoma is uncommon but any organ can be involved.. Extramedullary haematopoiesis is the production of red cells outside the medullary cavity in response to failure of erythrogenesis in bone marrow which can occur due to many diseases with thalassaemia and myelofibrosis being most common. We present a case of a 21 year old patient who underwent PET-CT scan for the staging of Hodgkin lymphoma and revealed co-existing extramedullary haematopoiesis secondary to known thalassaemia.


Subject(s)
Hematopoiesis, Extramedullary/physiology , Hodgkin Disease , Positron Emission Tomography Computed Tomography , beta-Thalassemia , Adult , Hodgkin Disease/diagnostic imaging , Hodgkin Disease/physiopathology , Humans , Male , Torso/diagnostic imaging , Young Adult , beta-Thalassemia/diagnostic imaging , beta-Thalassemia/physiopathology
7.
Osteoporos Int ; 29(1): 237-241, 2018 01.
Article in English | MEDLINE | ID: mdl-29071359

ABSTRACT

In fibrous dysplasia/McCune-Albright syndrome (FD/MAS), bone and bone marrow are, to varying degrees, replaced by fibro-osseous tissue typically devoid of hematopoietic marrow. Despite the extensive marrow replacement in severely affected patients, bone marrow failure is not commonly associated with FD/MAS. We present a 14-year-old girl with FD/MAS, who developed pancytopenia and extramedullary hematopoiesis (EMH) with no identified cause, in the setting of iatrogenic thyrotoxicosis and hyperparathyroidism. Pancytopenia, requiring monthly blood transfusions, persisted despite multiple strategies to correct these endocrinopathies. Due to worsening painful splenomegaly, likely as a result of sequestration, splenectomy was performed. Following splenectomy, pancytopenia resolved and patient has since been transfusion-independent. We report the first detailed case of bone marrow failure and EMH in FD/MAS. The etiology of marrow failure is likely multifactorial and related to the loss of marrow reserve due to extensive polyostotic FD, exacerbated by iatrogenic thyrotoxicosis and hyperparathyroidism. Mini Abstract: A patient with fibrous dysplasia developed bone marrow failure and extramedullary hematopoiesis. The etiology likely involved loss of hematopoetic marrow space and uncontrolled endocrinopathies. Splenectomy was therapeutic.


Subject(s)
Anemia, Aplastic/etiology , Bone Marrow Diseases/etiology , Fibrous Dysplasia, Polyostotic/complications , Hematopoiesis, Extramedullary/physiology , Hemoglobinuria, Paroxysmal/etiology , Adolescent , Anemia, Aplastic/pathology , Anemia, Aplastic/surgery , Biopsy , Bone Marrow/pathology , Bone Marrow Diseases/pathology , Bone Marrow Diseases/surgery , Bone Marrow Failure Disorders , Female , Fibrous Dysplasia, Polyostotic/diagnostic imaging , Fibrous Dysplasia, Polyostotic/physiopathology , Hemoglobinuria, Paroxysmal/pathology , Hemoglobinuria, Paroxysmal/surgery , Humans , Liver/pathology , Pancytopenia/etiology , Pancytopenia/surgery , Radiography , Splenectomy
8.
Eur Heart J ; 38(6): 425-432, 2017 02 07.
Article in English | MEDLINE | ID: mdl-27357356

ABSTRACT

Aims: Experimental work posits that acute ischaemic events trigger haematopoietic activity, driving monocytosis, and atherogenesis. Considering the chronic low-grade inflammatory state in atherosclerosis, we hypothesized that haematopoietic hyperactivity is a persistent feature in cardiovascular disease (CVD). Therefore, we aimed to assess the activity of haematopoietic organs and haematopoietic stem and progenitor cells (HSPCs) in humans. Methods and results: First, we performed 18F-fluorodeoxyglucose positron emission tomographic (18F-FDG PET) imaging in 26 patients with stable atherosclerotic CVD (ischaemic event >12 months ago), and 25 matched controls. In splenic tissue, 18F-FDG uptake was 2.68 ± 0.65 in CVD patients vs. 1.75 ± 0.54 in controls (1.6-fold higher; P< 0.001), and in bone marrow 3.20 ± 0.76 vs. 2.72 ± 0.46 (1.2-fold higher; P = 0.003), closely related to LDL cholesterol levels (LDLc, r = 0.72). Subsequently, we determined progenitor potential of HSPCs harvested from 18 patients with known atherosclerotic CVD and 30 matched controls; both groups were selected from a cohort of cancer patients undergoing autologous stem cell transplantation. In CVD patients, the normalized progenitor potential, expressed as the number of colony-forming units-granulocyte/monocyte (CFU-GM) colonies/CD34+ cell, was 1.6-fold higher compared with matched controls (P < 0.001). Finally, we assessed the effects of native and oxidized lipoproteins on HSPCs harvested from healthy donors in vitro. Haematopoietic stem and progenitor cells displayed a 1.5-fold increased CFU-GM capacity in co-culture with oxidized LDL in vitro (P = 0.002), which was inhibited by blocking oxidized phospholipids via E06 (P = 0.001). Conclusion: Collectively, these findings strengthen the case for a chronically affected haematopoietic system, potentially driving the low-grade inflammatory state in patients with atherosclerosis.


Subject(s)
Coronary Artery Disease/pathology , Hematopoiesis/physiology , Hematopoietic Stem Cells/physiology , Vascular Calcification/pathology , Bone Marrow/physiology , Case-Control Studies , Cells, Cultured , Cholesterol, LDL/physiology , Female , Fluorodeoxyglucose F18 , Hematopoiesis, Extramedullary/physiology , Humans , Male , Middle Aged , Positron Emission Tomography Computed Tomography , Radiopharmaceuticals , Risk Factors
9.
J Formos Med Assoc ; 117(12): 1108-1114, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29317181

ABSTRACT

BACKGROUND/PURPOSE: The thyroid gland is an uncommon site for the presence of extramedullary hematopoiesis (EMH). We report the cases of four Taiwanese women with hematopoietic elements on the smears of thyroid fine needle aspiration (FNA) samples and compare the findings with those of previously reported cases. METHODS: We retrieved the data of cases in which thyroid FNA performed between January 2000 and December 2016. The clinical manifestations, laboratory data, and image findings of cases with thyroid EMH were reported. A review of English literature was performed, and the reported cases were compared with our series. RESULTS: During the study period, 63,361 specimens of thyroid FNAs were identified. Four specimens contained hematopoietic elements from four women. Ultrasound study revealed calcifications in three patients. A review of English literature yielded 18 reports of thyroid EMH involving 29 patients. Twenty-six patients were women, and three were men. Nodule calcifications were found in 13 patients. Four patients had primary myelofibrosis, and one had chronic anemia. None had a thyroid malignancy. CONCLUSION: Four patients were noted to have bone marrow elements on the smears among 63,361 thyroid FNA samples. These four Taiwanese women presented with nodular goiter and foci of hematopoietic elements by thyroid FNA without any chronic disease of hematopoietic tissue or any evidence of a thyroid malignancy.


Subject(s)
Hematopoiesis, Extramedullary/physiology , Thyroid Gland/pathology , Thyroid Gland/physiopathology , Aged , Biopsy, Fine-Needle , Diagnosis, Differential , Female , Humans , Middle Aged , Thyroid Nodule/complications
10.
Int J Mol Sci ; 19(3)2018 Mar 18.
Article in English | MEDLINE | ID: mdl-29562644

ABSTRACT

Myelofibrosis (MF) is a clinical manifestation of chronic BCR-ABL1-negative chronic myeloproliferative neoplasms. Splenomegaly is one of the major clinical manifestations of MF and is directly linked to splenic extramedullary hematopoiesis (EMH). EMH is associated with abnormal trafficking patterns of clonal hematopoietic cells due to the dysregulated bone marrow (BM) microenvironment leading to progressive splenomegaly. Several recent data have emphasized the role of several cytokines for splenic EMH. Alteration of CXCL12/CXCR4 pathway could also lead to splenic EMH by migrated clonal hematopoietic cells from BM to the spleen. Moreover, low Gata1 expression was found to be significantly associated with the EMH. Several gene mutations were found to be associated with significant splenomegaly in MF. In recent data, JAK2V617F homozygous mutation was associated with a larger spleen size. In other data, CALR mutations in MF were signigicantly associated with longer larger splenomegaly-free survivals than others. In addition, MF patients with ≥1 mutations in AZXL1, EZH1 or IDH1/2 had significantly low spleen reduction response in ruxolitinib treatment. Developments of JAK inhibitors, such as ruxolitinib, pacritinib, momelotinib, and febratinib enabled the effective management in MF patients. Especially, significant spleen reduction responses of the drugs were demonstrated in several randomized clinical studies, although those could not eradicate allele burdens of MF.


Subject(s)
Hematopoiesis, Extramedullary/physiology , Janus Kinases/genetics , Primary Myelofibrosis/genetics , Primary Myelofibrosis/pathology , Splenomegaly/genetics , Bone Marrow/pathology , Cell Movement , Chemokine CXCL12/metabolism , Hematopoiesis, Extramedullary/drug effects , Humans , Janus Kinase Inhibitors/pharmacology , Janus Kinase Inhibitors/therapeutic use , Mutation , Primary Myelofibrosis/drug therapy , Primary Myelofibrosis/physiopathology , Randomized Controlled Trials as Topic , Receptors, CXCR4/metabolism , Spleen/pathology , Splenomegaly/drug therapy , Splenomegaly/physiopathology
11.
Stem Cells ; 34(2): 431-44, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26418893

ABSTRACT

Adult-repopulating hematopoietic stem cells (HSCs) emerge in low numbers in the midgestation mouse embryo from a subset of arterial endothelium, through an endothelial-to-hematopoietic transition. HSC-producing arterial hemogenic endothelium relies on the establishment of embryonic blood flow and arterial identity, and requires ß-catenin signaling. Specified prior to and during the formation of these initial HSCs are thousands of yolk sac-derived erythro-myeloid progenitors (EMPs). EMPs ensure embryonic survival prior to the establishment of a permanent hematopoietic system, and provide subsets of long-lived tissue macrophages. While an endothelial origin for these HSC-independent definitive progenitors is also accepted, the spatial location and temporal output of yolk sac hemogenic endothelium over developmental time remain undefined. We performed a spatiotemporal analysis of EMP emergence, and document the morphological steps of the endothelial-to-hematopoietic transition. Emergence of rounded EMPs from polygonal clusters of Kit(+) cells initiates prior to the establishment of arborized arterial and venous vasculature in the yolk sac. Interestingly, Kit(+) polygonal clusters are detected in both arterial and venous vessels after remodeling. To determine whether there are similar mechanisms regulating the specification of EMPs with other angiogenic signals regulating adult-repopulating HSCs, we investigated the role of embryonic blood flow and Wnt/ß-catenin signaling during EMP emergence. In embryos lacking a functional circulation, rounded Kit(+) EMPs still fully emerge from unremodeled yolk sac vasculature. In contrast, canonical Wnt signaling appears to be a common mechanism regulating hematopoietic emergence from hemogenic endothelium. These data illustrate the heterogeneity in hematopoietic output and spatiotemporal regulation of primary embryonic hemogenic endothelium.


Subject(s)
Endothelium, Vascular/metabolism , Hematopoiesis, Extramedullary/physiology , Hematopoietic Stem Cells/metabolism , Wnt Proteins/metabolism , Yolk Sac/metabolism , Animals , Endothelium, Vascular/cytology , Hematopoietic Stem Cells/cytology , Mice , Mice, Transgenic , Yolk Sac/blood supply , Yolk Sac/cytology
12.
Eur Heart J ; 37(14): 1113-21, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-26869607

ABSTRACT

Atherosclerosis, the major underlying cause of cardiovascular disease, is characterized by a lipid-driven infiltration of inflammatory cells in large and medium arteries. Increased production and activation of monocytes, neutrophils, and platelets, driven by hypercholesterolaemia and defective high-density lipoproteins-mediated cholesterol efflux, tissue necrosis and cytokine production after myocardial infarction, or metabolic abnormalities associated with diabetes, contribute to atherogenesis and athero-thrombosis. This suggests that in addition to traditional approaches of low-density lipoproteins lowering and anti-platelet drugs, therapies directed at abnormal haematopoiesis, including anti-inflammatory agents, drugs that suppress myelopoiesis, and excessive platelet production, rHDL infusions and anti-obesity and anti-diabetic agents, may help to prevent athero-thrombosis.


Subject(s)
Atherosclerosis/physiopathology , Hematopoiesis/physiology , Thrombosis/physiopathology , Blood Platelets/physiology , Cholesterol/metabolism , Hematopoiesis, Extramedullary/physiology , Hematopoietic Stem Cells/physiology , Humans , Leukocytosis/complications , Leukocytosis/physiopathology , Metabolic Diseases/complications , Metabolic Diseases/physiopathology , Monocytes/physiology , Myelopoiesis/physiology , Myeloproliferative Disorders/complications , Myeloproliferative Disorders/physiopathology , Neutrophils/physiology , Risk Factors , Thrombopoiesis/physiology
13.
J Biol Chem ; 289(14): 9795-810, 2014 Apr 04.
Article in English | MEDLINE | ID: mdl-24509850

ABSTRACT

Abnormal blood cell production is associated with chronic kidney disease (CKD) and cardiovascular disease (CVD). Bone-derived FGF-23 (fibroblast growth factor-23) regulates phosphate homeostasis and bone mineralization. Genetic deletion of Fgf-23 in mice (Fgf-23(-/-)) results in hypervitaminosis D, abnormal mineral metabolism, and reduced lymphatic organ size. Elevated FGF-23 levels are linked to CKD and greater risk of CVD, left ventricular hypertrophy, and mortality in dialysis patients. However, whether FGF-23 is involved in the regulation of erythropoiesis is unknown. Here we report that loss of FGF-23 results in increased hematopoietic stem cell frequency associated with increased erythropoiesis in peripheral blood and bone marrow in young adult mice. In particular, these hematopoietic changes are also detected in fetal livers, suggesting that they are not the result of altered bone marrow niche alone. Most importantly, administration of FGF-23 in wild-type mice results in a rapid decrease in erythropoiesis. Finally, we show that the effect of FGF-23 on erythropoiesis is independent of the high vitamin D levels in these mice. Our studies suggest a novel role for FGF-23 in erythrocyte production and differentiation and suggest that elevated FGF-23 levels contribute to the pathogenesis of anemia in patients with CKD and CVD.


Subject(s)
Bone Marrow/metabolism , Cell Differentiation/physiology , Erythrocytes/metabolism , Erythropoiesis/physiology , Fetus/metabolism , Fibroblast Growth Factors/metabolism , Anemia/genetics , Anemia/metabolism , Animals , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/therapy , Erythrocytes/cytology , Fetus/cytology , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/genetics , Hematopoiesis, Extramedullary/physiology , Humans , Liver/cytology , Liver/embryology , Liver/metabolism , Mice , Mice, Knockout , Renal Dialysis/adverse effects , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/therapy , Risk Factors , Vitamin D/genetics , Vitamin D/metabolism
14.
Br J Haematol ; 168(6): 891-901, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25403101

ABSTRACT

Effective erythropoiesis requires an appropriate supply of iron and mechanisms regulating iron homeostasis and erythropoiesis are intrinsically linked. Iron dysregulation, typified by iron-deficiency anaemia and iron overload, is common in many clinical conditions and impacts the health of up to 30% of the world's population. The proteins transmembrane protease, serine 6 (TMPRSS6; also termed matriptase-2), HFE and transferrin receptor 2 (TFR2) play important and opposing roles in systemic iron homeostasis, by regulating expression of the iron regulatory hormone hepcidin. We have performed a systematic analysis of mice deficient in these three proteins and show that TMPRSS6 predominates over HFE and TFR2 in hepcidin regulation. The phenotype of mice lacking TMPRSS6 and TFR2 is characterized by severe anaemia and extramedullary haematopoiesis in the spleen. Stress erythropoiesis in these mice results in increased expression of the newly identified erythroid iron regulator erythroferrone, which does not appear to overcome the hepcidin overproduction mediated by loss of TMPRSS6. Extended analysis reveals that TFR2 plays an important role in erythroid cells, where it is involved in terminal erythroblast differentiation and the regulation of erythropoietin. In conclusion, we have identified an essential role for TFR2 in erythropoiesis that may provide new targets for the treatment of anaemia.


Subject(s)
Anemia, Iron-Deficiency/blood , Erythropoiesis/physiology , Receptors, Transferrin/physiology , Anemia, Iron-Deficiency/metabolism , Animals , Cell Differentiation/physiology , Erythroid Cells/pathology , Erythropoietin/biosynthesis , Hematopoiesis, Extramedullary/physiology , Hemochromatosis Protein , Hepcidins/metabolism , Histocompatibility Antigens Class I/blood , Histocompatibility Antigens Class I/physiology , Kidney/metabolism , Liver/metabolism , Male , Membrane Proteins/blood , Membrane Proteins/deficiency , Membrane Proteins/physiology , Mice , Mice, Knockout , Receptors, Erythropoietin/metabolism , Receptors, Transferrin/blood , Receptors, Transferrin/deficiency , Serine Endopeptidases/blood , Serine Endopeptidases/deficiency , Serine Endopeptidases/physiology , Splenomegaly/blood
15.
Blood ; 121(8): 1436-45, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23264599

ABSTRACT

Erythropoiesis must be tightly balanced to guarantee adequate oxygen delivery to all tissues in the body. This process relies predominantly on the hormone erythropoietin (EPO) and its transcription factor hypoxia inducible factor (HIF). Accumulating evidence suggests that oxygen-sensitive prolyl hydroxylases (PHDs) are important regulators of this entire system. Here, we describe a novel mouse line with conditional PHD2 inactivation (cKO P2) in renal EPO producing cells, neurons, and astrocytes that displayed excessive erythrocytosis because of severe overproduction of EPO, exclusively driven by HIF-2α. In contrast, HIF-1α served as a protective factor, ensuring survival of cKO P2 mice with HCT values up to 86%. Using different genetic approaches, we show that simultaneous inactivation of PHD2 and HIF-1α resulted in a drastic PHD3 reduction with consequent overexpression of HIF-2α-related genes, neurodegeneration, and lethality. Taken together, our results demonstrate for the first time that conditional loss of PHD2 in mice leads to HIF-2α-dependent erythrocytosis, whereas HIF-1α protects these mice, providing a platform for developing new treatments of EPO-related disorders, such as anemia.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Hematopoiesis, Extramedullary/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Polycythemia/genetics , Procollagen-Proline Dioxygenase/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Brain/physiology , Cells, Cultured , Erythropoietin/genetics , Erythropoietin/metabolism , Female , Fibroblasts/cytology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases , Keratinocytes/cytology , Kidney/cytology , Kidney/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Polycythemia/metabolism , Polycythemia/pathology , Procollagen-Proline Dioxygenase/metabolism , Severity of Illness Index , Thrombocytopenia/genetics , Thrombocytopenia/metabolism , Thrombocytopenia/pathology
16.
Duodecim ; 131(7): 671-5, 2015.
Article in Fi | MEDLINE | ID: mdl-26233985

ABSTRACT

Sometimes correct diagnoses is reached after many years and even after decades. Our patient had for decades suffered from a hemolytic disease, life-threatening, metastatic cancer at the age of almost 90 years was also suspected. The patient was finally diagnosed as having mild hereditary spherocytosis and the associated paraspinal extramedullar hematopoiesis as well as an osteoporotic vertebral fracture caused by osteoporosis.


Subject(s)
Hematopoiesis, Extramedullary/physiology , Osteoporotic Fractures/diagnosis , Spherocytosis, Hereditary/diagnosis , Spinal Fractures/diagnosis , Aged, 80 and over , Diagnosis, Differential , Humans , Male
17.
Blood ; 119(21): 4971-80, 2012 May 24.
Article in English | MEDLINE | ID: mdl-22490334

ABSTRACT

The interactions between hematopoietic cells and the bone marrow (BM) microenvironment play a critical role in normal and malignant hematopoiesis and drug resistance. These interactions within the BM niche are unique and could be important for developing new therapies. Here, we describe the development of extramedullary bone and bone marrow using human mesenchymal stromal cells and endothelial colony-forming cells implanted subcutaneously into immunodeficient mice. We demonstrate the engraftment of human normal and leukemic cells engraft into the human extramedullary bone marrow. When normal hematopoietic cells are engrafted into the model, only discrete areas of the BM are hypoxic, whereas leukemia engraftment results in widespread severe hypoxia, just as recently reported by us in human leukemias. Importantly, the hematopoietic cell engraftment could be altered by genetical manipulation of the bone marrow microenvironment: Extramedullary bone marrow in which hypoxia-inducible factor 1α was knocked down in mesenchymal stromal cells by lentiviral transfer of short hairpin RNA showed significant reduction (50% ± 6%; P = .0006) in human leukemic cell engraftment. These results highlight the potential of a novel in vivo model of human BM microenvironment that can be genetically modified. The model could be useful for the study of leukemia biology and for the development of novel therapeutic modalities aimed at modifying the hematopoietic microenvironment.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Transplantation/methods , Cellular Microenvironment/physiology , Hematopoiesis, Extramedullary/physiology , Transplantation, Heterotopic , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/physiology , Bone Marrow Transplantation/physiology , Cells, Cultured , Cellular Microenvironment/genetics , Hematopoiesis, Extramedullary/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin Receptor Common gamma Subunit/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Models, Animal , Osteogenesis/genetics , Osteogenesis/physiology , Species Specificity
18.
Mol Cell Proteomics ; 11(10): 1024-35, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22822186

ABSTRACT

Menstruation is the expulsion of the endometrial lining of the uterus following a nearly month long preparation for embryo implantation and pregnancy. Increasingly, the health of the endometrium is being recognized as a critical factor in female fertility, and proteomes and transcriptomes from endometrial biopsies at different stages of the menstrual cycle have been studied for both diagnostic and therapeutic purposes (1 Kao, L. C., et al. 2003 Endocrinology 144, 2870-2881; Strowitzki, Tet al. 2006 Hum. Reprod. Update 12, 617-630; DeSouza, L., et al. 2005 Proteomics 5, 270-281). Disorders of the uterus ranging from benign to malignant tumors, as well as endometriosis, can cause abnormal menstrual bleeding and are frequently diagnosed through endometrial biopsy (Strowitzki, Tet al. 2006 Hum. Reprod. Update 12, 617-630; Ferenczy, A. 2003 Maturitas 45, 1-14). Yet the proteome of menstrual blood, an easily available noninvasive source of endometrial tissue, has yet to be examined for possible causes or diagnoses of infertility or endometrial pathology. This study employed five different methods to define the menstrual blood proteome. A total of 1061 proteins were identified, 361 were found by at least two methods and 678 were identified by at least two peptides. When the menstrual blood proteome was compared with those of circulating blood (1774 proteins) and vaginal fluid (823 proteins), 385 proteins were found unique to menstrual blood. Gene ontology analysis and evaluation of these specific menstrual blood proteins identified pathways consistent with the processes of the normal endometrial cycle. Several of the proteins unique to menstrual blood suggest that extramedullary uterine hematopoiesis or parenchymal hemoglobin synthesis may be occurring in late endometrial tissue. The establishment of a normal menstrual blood proteome is necessary for the evaluation of its usefulness as a diagnostic tool for infertility and uterine pathologies. Identification of unique menstrual blood proteins should aid the forensic community in distinguishing menstrual blood from circulating blood.


Subject(s)
Body Fluids/chemistry , Endometrium/metabolism , Menstrual Cycle/blood , Menstruation/blood , Proteome/analysis , Adult , Chromatography, High Pressure Liquid , Endometrium/chemistry , Female , Hematopoiesis, Extramedullary/physiology , Hemoglobins/biosynthesis , Humans , Middle Aged , Proteomics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Vagina/chemistry , Vagina/metabolism
19.
Rev Chil Pediatr ; 85(2): 203-6, 2014 Apr.
Article in Spanish | MEDLINE | ID: mdl-25697209

ABSTRACT

UNLABELLED: Extramedullary hematopoiesis (EMH) is defined as the formation and development of erythrocytic, granulocytic and megakaryocytic cell lines outside the bone marrow; it is a rare occurrence, which is limited to case reports. OBJECTIVE: To describe a case of renal EMH secondary to chronic hypoxia. CASE REPORT: The patient is a 16 year old teenager who developed chronic hypoxia, central cyanosis, clubbing and lives more than 2,500 m above sea level. Chronic hypoxia is probably secondary to pulmonary hypertension, neither detected nor previously treated. The Consultation was due to a mass in the left upper quadrant for the last three months. An Echocardiogram showed pulmonary hypertension that could not be assessed by hemodynamic parameters. A computed tomography showed bilateral left renal pelvic masses. The hemogram showed polycythemia, so bone marrow aspiration was performed, which rule out malignant pathology. A biopsy of the renal mass reported EMH with three hematopoietic cell lines. CONCLUSION: This EMH case is a response to chronic hypoxia rather than to a bone marrow lesion as it has usually been described.


Subject(s)
Hematopoiesis, Extramedullary/physiology , Hypertension, Pulmonary/complications , Hypoxia/complications , Adolescent , Biopsy , Chronic Disease , Humans , Hypoxia/etiology , Male , Tomography, X-Ray Computed
20.
Blood ; 117(23): 6132-4, 2011 Jun 09.
Article in English | MEDLINE | ID: mdl-21505195

ABSTRACT

Hematopoietic cell clusters associated with the midgestation mouse aorta, umbilical and vitelline arteries play a pivotal role in the formation of the adult blood system. Both genetic and live-imaging data indicate that definitive hematopoietic progenitor/stem cells (visualized as clusters) are generated from hemogenic endothelium. A 3-dimensional (3-D) whole embryo immunostaining and imaging technique has allowed quantitation and cartographic mapping of intravascular hematopoietic clusters. During this period the vitelline artery is most extensively remodeled, and several reports have suggested that vitelline remodeling leads to extravascular hematopoietic cluster emergence. Whether the earliest definitive progenitors/stem cells are intra or extra vascular could influence the process by which these cells migrate to the next hematopoietic territory, the fetal liver. Hence, by 3-D imaging we more closely examined hematopoietic clusters in the vitelline and associated connected small vessels and show here that hematopoietic clusters (particularly large clusters) are intravascular during the period of vascular remodeling.


Subject(s)
Cell Movement/physiology , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Hematopoiesis, Extramedullary/physiology , Hematopoietic Stem Cells/cytology , Imaging, Three-Dimensional , Animals , Hematopoietic Stem Cells/metabolism , Mice , Mice, Transgenic , Microscopy, Confocal/methods
SELECTION OF CITATIONS
SEARCH DETAIL