243 results on '"Rivella, S."'
Search Results
152. Intestinal HIF2α promotes tissue-iron accumulation in disorders of iron overload with anemia.
- Author
-
Anderson ER, Taylor M, Xue X, Ramakrishnan SK, Martin A, Xie L, Bredell BX, Gardenghi S, Rivella S, and Shah YM
- Subjects
- Analysis of Variance, Animals, Blotting, Western, Ferrocyanides, Iron Overload metabolism, Luciferases, Mice, Real-Time Polymerase Chain Reaction, Transcription Factors metabolism, beta-Thalassemia metabolism, Basic Helix-Loop-Helix Transcription Factors metabolism, Intestinal Mucosa metabolism, Iron Overload etiology, beta-Thalassemia complications
- Abstract
Several distinct congenital disorders can lead to tissue-iron overload with anemia. Repeated blood transfusions are one of the major causes of iron overload in several of these disorders, including β-thalassemia major, which is characterized by a defective β-globin gene. In this state, hyperabsorption of iron is also observed and can significantly contribute to iron overload. In β-thalassemia intermedia, which does not require blood transfusion for survival, hyperabsorption of iron is the leading cause of iron overload. The mechanism of increased iron absorption in β-thalassemia is unclear. We definitively demonstrate, using genetic mouse models, that intestinal hypoxia-inducible factor-2α (HIF2α) and divalent metal transporter-1 (DMT1) are activated early in the pathogenesis of β-thalassemia and are essential for excess iron accumulation in mouse models of β-thalassemia. Moreover, thalassemic mice with established iron overload had significant improvement in tissue-iron levels and anemia following disruption of intestinal HIF2α. In addition to repeated blood transfusions and increased iron absorption, chronic hemolysis is the major cause of tissue-iron accumulation in anemic iron-overload disorders caused by hemolytic anemia. Mechanistic studies in a hemolytic anemia mouse model demonstrated that loss of intestinal HIF2α/DMT1 signaling led to decreased tissue-iron accumulation in the liver without worsening the anemia. These data demonstrate that dysregulation of intestinal hypoxia and HIF2α signaling is critical for progressive iron overload in β-thalassemia and may be a novel therapeutic target in several anemic iron-overload disorders.
- Published
- 2013
- Full Text
- View/download PDF
153. Isocitrate ameliorates anemia by suppressing the erythroid iron restriction response.
- Author
-
Richardson CL, Delehanty LL, Bullock GC, Rival CM, Tung KS, Kimpel DL, Gardenghi S, Rivella S, and Goldfarb AN
- Subjects
- Aconitate Hydratase metabolism, Anemia metabolism, Anemia pathology, Animals, Cells, Cultured, Erythroid Cells enzymology, Female, Humans, Interferon-gamma physiology, Isocitrates therapeutic use, Protein Kinase C metabolism, Proto-Oncogene Proteins metabolism, Rats, Rats, Inbred Lew, Signal Transduction, Trans-Activators metabolism, Transcriptional Activation, Anemia drug therapy, Erythroid Cells drug effects, Erythropoiesis drug effects, Iron Deficiencies, Isocitrates pharmacology
- Abstract
The unique sensitivity of early red cell progenitors to iron deprivation, known as the erythroid iron restriction response, serves as a basis for human anemias globally. This response impairs erythropoietin-driven erythropoiesis and underlies erythropoietic repression in iron deficiency anemia. Mechanistically, the erythroid iron restriction response results from inactivation of aconitase enzymes and can be suppressed by providing the aconitase product isocitrate. Recent studies have implicated the erythroid iron restriction response in anemia of chronic disease and inflammation (ACDI), offering new therapeutic avenues for a major clinical problem; however, inflammatory signals may also directly repress erythropoiesis in ACDI. Here, we show that suppression of the erythroid iron restriction response by isocitrate administration corrected anemia and erythropoietic defects in rats with ACDI. In vitro studies demonstrated that erythroid repression by inflammatory signaling is potently modulated by the erythroid iron restriction response in a kinase-dependent pathway involving induction of the erythroid-inhibitory transcription factor PU.1. These results reveal the integration of iron and inflammatory inputs in a therapeutically tractable erythropoietic regulatory circuit.
- Published
- 2013
- Full Text
- View/download PDF
154. Combining gene therapy and fetal hemoglobin induction for treatment of β-thalassemia.
- Author
-
Breda L, Rivella S, Zuccato C, and Gambari R
- Subjects
- Antisickling Agents therapeutic use, Azacitidine analogs & derivatives, Azacitidine therapeutic use, Decitabine, Fetal Hemoglobin genetics, Genetic Therapy, Genetic Vectors genetics, Genetic Vectors metabolism, Humans, Hydroxyurea therapeutic use, Lentivirus genetics, Retroviridae genetics, Thalidomide therapeutic use, beta-Globins genetics, Fetal Hemoglobin metabolism, beta-Globins metabolism, beta-Thalassemia therapy
- Abstract
β-thalassemias are caused by nearly 300 mutations of the β-globin gene, leading to a low or absent production of adult hemoglobin (HbA). Two major therapeutic approaches have recently been proposed: gene therapy and induction of fetal hemoglobin (HbF) with the objective of achieving clinically relevant levels of Hbs. The objective of this article is to describe the development of therapeutic strategies based on a combination of gene therapy and induction of HbFs. An increase of β-globin gene expression in β-thalassemia cells can be achieved by gene therapy, although de novo production of clinically relevant levels of adult Hb may be difficult to obtain. On the other hand, an increased production of HbF is beneficial in β-thalassemia. The combination of gene therapy and HbF induction appears to be a pertinent strategy to achieve clinically relevant results.
- Published
- 2013
- Full Text
- View/download PDF
155. Reducing TMPRSS6 ameliorates hemochromatosis and β-thalassemia in mice.
- Author
-
Guo S, Casu C, Gardenghi S, Booten S, Aghajan M, Peralta R, Watt A, Freier S, Monia BP, and Rivella S
- Subjects
- Animals, Antimicrobial Cationic Peptides genetics, Antimicrobial Cationic Peptides metabolism, Cells, Cultured, Female, Gene Knockdown Techniques, Hemochromatosis blood, Hemochromatosis genetics, Hemochromatosis Protein, Hepatocytes metabolism, Hepcidins, Histocompatibility Antigens Class I genetics, Histocompatibility Antigens Class I metabolism, Iron blood, Iron metabolism, Liver metabolism, Male, Membrane Proteins metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Oligonucleotides, Antisense genetics, RNA, Messenger genetics, RNA, Messenger metabolism, Serine Endopeptidases metabolism, Spleen metabolism, Spleen pathology, Transferrin metabolism, beta-Thalassemia blood, beta-Thalassemia genetics, Hemochromatosis therapy, Membrane Proteins genetics, Serine Endopeptidases genetics, beta-Thalassemia therapy
- Abstract
β-Thalassemia and HFE-related hemochromatosis are 2 of the most frequently inherited disorders worldwide. Both disorders are characterized by low levels of hepcidin (HAMP), the hormone that regulates iron absorption. As a consequence, patients affected by these disorders exhibit iron overload, which is the main cause of morbidity and mortality. HAMP expression is controlled by activation of the SMAD1,5,8/SMAD4 complex. TMPRSS6 is a serine protease that reduces SMAD activation and blocks HAMP expression. We identified second generation antisense oligonucleotides (ASOs) targeting mouse Tmprss6. ASO treatment in mice affected by hemochromatosis (Hfe(-/-)) significantly decreased serum iron, transferrin saturation and liver iron accumulation. Furthermore, ASO treatment of mice affected by β-thalassemia (HBB(th3/+) mice, referred to hereafter as th3/+ mice) decreased the formation of insoluble membrane-bound globins, ROS, and apoptosis, and improved anemia. These animals also exhibited lower erythropoietin levels, a significant amelioration of ineffective erythropoiesis (IE) and splenomegaly, and an increase in total hemoglobin levels. These data suggest that ASOs targeting Tmprss6 could be beneficial in individuals with hemochromatosis, β-thalassemia, and related disorders.
- Published
- 2013
- Full Text
- View/download PDF
156. Gene therapy for hemoglobinopathies: progress and challenges.
- Author
-
Dong A, Rivella S, and Breda L
- Subjects
- Animals, Clinical Trials as Topic, Disease Models, Animal, Fetal Hemoglobin metabolism, Humans, Phenotype, Genetic Therapy, Hemoglobinopathies genetics, Hemoglobinopathies therapy
- Abstract
Hemoglobinopathies are genetic inherited conditions that originate from the lack or malfunction of the hemoglobin (Hb) protein. Sickle cell disease (SCD) and thalassemia are the most common forms of these conditions. The severe anemia combined with complications that arise in the most affected patients raises the necessity for a cure to restore hemoglobin function. The current routine therapies for these conditions, namely transfusion and iron chelation, have significantly improved the quality of life in patients over the years, but still fail to address the underlying cause of the diseases. A curative option, allogeneic bone marrow transplantation is available, but limited by the availability of suitable donors and graft-vs-host disease. Gene therapy offers an alternative approach to cure patients with hemoglobinopathies and aims at the direct recovery of the hemoglobin function via globin gene transfer. In the last 2 decades, gene transfer tools based on lentiviral vector development have been significantly improved and proven curative in several animal models for SCD and thalassemia. As a result, clinical trials are in progress and 1 patient has been successfully treated with this approach. However, there are still frontiers to explore that might improve this approach: the stoichiometry between the transgenic hemoglobin and endogenous hemoglobin with respect to the different globin genetic mutations; donor cell sourcing, such as the use of induced pluripotent stem cells (iPSCs); and the use of safer gene insertion methods to prevent oncogenesis. With this review we will provide insights about (1) the different lentiviral gene therapy approaches in mouse models and human cells; (2) current and planned clinical trials; (3) hurdles to overcome for clinical trials, such as myeloablation toxicity, insertional oncogenesis, and high vector expression; and (4) future perspectives for gene therapy, including safe harbors and iPSCs technology., (Copyright © 2013 Mosby, Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
157. Macrophages support pathological erythropoiesis in polycythemia vera and β-thalassemia.
- Author
-
Ramos P, Casu C, Gardenghi S, Breda L, Crielaard BJ, Guy E, Marongiu MF, Gupta R, Levine RL, Abdel-Wahab O, Ebert BL, Van Rooijen N, Ghaffari S, Grady RW, Giardina PJ, and Rivella S
- Subjects
- Animals, Clodronic Acid pharmacology, Disease Models, Animal, Erythrocyte Count, Erythropoiesis drug effects, Female, Hematocrit, Hemoglobins analysis, Humans, Macrophages drug effects, Male, Mice, Mice, Inbred C57BL, Reticulocytes physiology, Stress, Physiological physiology, Erythropoiesis physiology, Macrophages physiology, Polycythemia Vera physiopathology, beta-Thalassemia physiopathology
- Abstract
Regulation of erythropoiesis is achieved by the integration of distinct signals. Among them, macrophages are emerging as erythropoietin-complementary regulators of erythroid development, particularly under stress conditions. We investigated the contribution of macrophages to physiological and pathological conditions of enhanced erythropoiesis. We used mouse models of induced anemia, polycythemia vera and β-thalassemia in which macrophages were chemically depleted. Our data indicate that macrophages contribute decisively to recovery from induced anemia, as well as the pathological progression of polycythemia vera and β-thalassemia, by modulating erythroid proliferation and differentiation. We validated these observations in primary human cultures, showing a direct impact of macrophages on the proliferation and enucleation of erythroblasts from healthy individuals and patients with polycythemia vera or β-thalassemia. The contribution of macrophages to stress and pathological erythropoiesis, which we have termed stress erythropoiesis macrophage-supporting activity, may have therapeutic implications.
- Published
- 2013
- Full Text
- View/download PDF
158. The murine growth differentiation factor 15 is not essential for systemic iron homeostasis in phlebotomized mice.
- Author
-
Casanovas G, Vujić Spasic M, Casu C, Rivella S, Strelau J, Unsicker K, and Muckenthaler MU
- Subjects
- Animals, Bone Marrow metabolism, Erythrocyte Indices, Female, Growth Differentiation Factor 15 genetics, Hepcidins genetics, Hepcidins metabolism, Iron blood, Liver metabolism, Male, Mice, Mice, Knockout, Phlebotomy, RNA, Messenger genetics, RNA, Messenger metabolism, Spleen metabolism, Growth Differentiation Factor 15 metabolism, Homeostasis, Iron metabolism
- Abstract
In conditions of increased erythropoiesis, expression of hepcidin, the master regulator of systemic iron homeostasis, is decreased to allow for the release of iron into the blood stream from duodenal enterocytes and macrophages. It has been suggested that hepcidin suppression is controlled by growth differentiation factor 15 (GDF15), a member of the transforming growth factor-β superfamily of cytokines that is secreted from developing erythroblasts. In this study, we analyzed iron-related parameters in mice deficient for GDF15 under steady-state conditions and in response to increased erythropoietic activity induced by blood loss. We demonstrate that GDF15 suppresses the hepatic mRNA expression of some BMP/TGFβ target genes but not of hepcidin, and show that GDF15 is not required to balance iron homeostasis in response to blood loss.
- Published
- 2013
- Full Text
- View/download PDF
159. In vivo gene transfer strategies to achieve partial correction of von Willebrand disease.
- Author
-
Wang L, Rosenberg JB, De BP, Ferris B, Wang R, Rivella S, Kaminsky SM, and Crystal RG
- Subjects
- Animals, Dependovirus genetics, Genetic Vectors genetics, Immunohistochemistry, Liver pathology, Mice, Mice, Knockout, von Willebrand Diseases genetics, Genetic Therapy, von Willebrand Diseases therapy
- Abstract
von Willebrand disease (VWD), the most common hereditary coagulation disorder, results from mutations in the 52-exon gene for von Willebrand factor (VWF), which encodes an 8.4-kB cDNA. Studies with VWF cDNA plasmids have demonstrated that in vivo gene transfer to the liver will correct the coagulation dysfunction in VWF(-/-) mice, but the correction is transient. To develop gene therapy for VWF that would mediate long-term expression of the VWF cDNA in liver, we first evaluated segmental pre-mRNA trans-splicing (SPTS) with two adeno-associated virus (AAV) serotype 8 vectors, each delivering one-half of the VWF cDNA. However, although the two vectors functioned well to generate VWF multimers after infection of cells in vitro, the efficiency of SPTS was insufficient to correct the VWF(-/-) mouse in vivo. As an alternative, we assessed the ability of a lentiviral vector to transfer the intact murine VWF cDNA in vivo directly to the neonatal liver of VWF(-/-) mice, using generation of VWF multimers, bleeding time, and bleeding volume as efficacy parameters. The VWF lentivirus generated VWF multimers and partially or completely corrected the coagulation defect on a persistent basis in 33% of the treated VWF-deficient mice. On the basis of the concept that partial persistent correction with gene transfer could be beneficial in VWD patients, these observations suggest that lentiviral delivery of VWF cDNA should be explored as a candidate for gene therapy in patients with a severe form of VWD.
- Published
- 2012
- Full Text
- View/download PDF
160. Do not super-excess me!
- Author
-
Rivella S
- Subjects
- Animals, Bortezomib, Female, Humans, Male, Alpha-Globulins metabolism, Antineoplastic Agents pharmacology, Boronic Acids pharmacology, Erythroid Precursor Cells metabolism, Proteasome Inhibitors, Proteolysis drug effects, Pyrazines pharmacology, Ubiquitination drug effects, beta-Thalassemia drug therapy
- Published
- 2012
- Full Text
- View/download PDF
161. Myeloid cell-derived hypoxia-inducible factor attenuates inflammation in unilateral ureteral obstruction-induced kidney injury.
- Author
-
Kobayashi H, Gilbert V, Liu Q, Kapitsinou PP, Unger TL, Rha J, Rivella S, Schlöndorff D, and Haase VH
- Subjects
- Acute Kidney Injury genetics, Animals, Basic Helix-Loop-Helix Transcription Factors deficiency, Disease Models, Animal, Fibrosis immunology, Fibrosis prevention & control, Humans, Hypoxia-Inducible Factor 1, alpha Subunit deficiency, Inflammation immunology, Inflammation pathology, Inflammation prevention & control, Mice, Mice, Knockout, Mice, Transgenic, Myeloid Cells metabolism, Primary Cell Culture, Ureteral Obstruction genetics, Acute Kidney Injury immunology, Acute Kidney Injury pathology, Basic Helix-Loop-Helix Transcription Factors physiology, Hypoxia-Inducible Factor 1, alpha Subunit physiology, Myeloid Cells immunology, Myeloid Cells pathology, Ureteral Obstruction immunology, Ureteral Obstruction pathology
- Abstract
Renal fibrosis and inflammation are associated with hypoxia, and tissue pO(2) plays a central role in modulating the progression of chronic kidney disease. Key mediators of cellular adaptation to hypoxia are hypoxia-inducible factor (HIF)-1 and -2. In the kidney, they are expressed in a cell type-specific manner; to what degree activation of each homolog modulates renal fibrogenesis and inflammation has not been established. To address this issue, we used Cre-loxP recombination to activate or to delete both Hif-1 and Hif-2 either globally or cell type specifically in myeloid cells. Global activation of Hif suppressed inflammation and fibrogenesis in mice subjected to unilateral ureteral obstruction, whereas activation of Hif in myeloid cells suppressed inflammation only. Suppression of inflammatory cell infiltration was associated with downregulation of CC chemokine receptors in renal macrophages. Conversely, global deletion or myeloid-specific inactivation of Hif promoted inflammation. Furthermore, prolonged hypoxia suppressed the expression of multiple inflammatory molecules in noninjured kidneys. Collectively, we provide experimental evidence that hypoxia and/or myeloid cell-specific HIF activation attenuates renal inflammation associated with chronic kidney injury.
- Published
- 2012
- Full Text
- View/download PDF
162. The role of ineffective erythropoiesis in non-transfusion-dependent thalassemia.
- Author
-
Rivella S
- Subjects
- Animals, Blood Transfusion, Humans, Iron metabolism, beta-Thalassemia metabolism, beta-Thalassemia pathology, Erythropoiesis physiology, beta-Thalassemia blood
- Abstract
Ineffective erythropoiesis is the hallmark of beta-thalassemia that triggers a cascade of compensatory mechanisms resulting in clinical sequelae such as erythroid marrow expansion, extramedullary hematopoiesis, splenomegaly, and increased gastrointestinal iron absorption. Recent studies have begun to shed light on the complex molecular mechanisms underlying ineffective erythropoiesis and the associated compensatory pathways; this new understanding may lead to the development of novel therapies. Increased or excessive activation of the Jak2/STAT5 pathway promotes unnecessary disproportionate proliferation of erythroid progenitors, while other factors suppress serum hepcidin levels leading to dysregulation of iron metabolism. Preclinical studies suggest that Jak inhibitors, hepcidin agonists, and exogenous transferrin may help to restore normal erythropoiesis and iron metabolism and reduce splenomegaly; however, further research is needed., (Copyright © 2012 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
163. Decreased hepcidin expression in murine β-thalassemia is associated with suppression of Bmp/Smad signaling.
- Author
-
Parrow NL, Gardenghi S, Ramos P, Casu C, Grady RW, Anderson ER, Shah YM, Li H, Ginzburg YZ, Fleming RE, and Rivella S
- Subjects
- Animals, Antimicrobial Cationic Peptides metabolism, Bone Morphogenetic Protein 6 genetics, Bone Morphogenetic Protein 6 metabolism, Bone Morphogenetic Proteins genetics, Bone Morphogenetic Proteins metabolism, Down-Regulation genetics, Gene Expression Regulation, Hepcidins, Iron metabolism, Iron Overload genetics, Iron Overload metabolism, Liver metabolism, Liver pathology, Mice, Mice, Inbred C57BL, Mice, Transgenic, Signal Transduction genetics, Signal Transduction physiology, Smad Proteins genetics, Smad Proteins metabolism, beta-Thalassemia metabolism, beta-Thalassemia pathology, Antimicrobial Cationic Peptides genetics, Bone Morphogenetic Proteins physiology, Smad Proteins physiology, beta-Thalassemia genetics
- Published
- 2012
- Full Text
- View/download PDF
164. Therapeutic hemoglobin levels after gene transfer in β-thalassemia mice and in hematopoietic cells of β-thalassemia and sickle cells disease patients.
- Author
-
Breda L, Casu C, Gardenghi S, Bianchi N, Cartegni L, Narla M, Yazdanbakhsh K, Musso M, Manwani D, Little J, Gardner LB, Kleinert DA, Prus E, Fibach E, Grady RW, Giardina PJ, Gambari R, and Rivella S
- Subjects
- Adult, Anemia, Sickle Cell blood, Anemia, Sickle Cell genetics, Animals, Ankyrins genetics, Antigens, CD34 metabolism, Base Sequence, Cell Differentiation genetics, Cell Line, Tumor, Cells, Cultured, Erythroid Precursor Cells metabolism, Gene Expression, Gene Transfer Techniques, Genetic Vectors genetics, Hemoglobins genetics, Humans, Insulator Elements genetics, Lentivirus genetics, Mice, Molecular Sequence Data, Mutation, NIH 3T3 Cells, beta-Globins genetics, beta-Thalassemia blood, beta-Thalassemia genetics, Anemia, Sickle Cell therapy, Genetic Therapy methods, Hemoglobins metabolism, beta-Thalassemia therapy
- Abstract
Preclinical and clinical studies demonstrate the feasibility of treating β-thalassemia and Sickle Cell Disease (SCD) by lentiviral-mediated transfer of the human β-globin gene. However, previous studies have not addressed whether the ability of lentiviral vectors to increase hemoglobin synthesis might vary in different patients.We generated lentiviral vectors carrying the human β-globin gene with and without an ankyrin insulator and compared their ability to induce hemoglobin synthesis in vitro and in thalassemic mice. We found that insertion of an ankyrin insulator leads to higher, potentially therapeutic levels of human β-globin through a novel mechanism that links the rate of transcription of the transgenic β-globin mRNA during erythroid differentiation with polysomal binding and efficient translation, as reported here for the first time. We also established a preclinical assay to test the ability of this novel vector to synthesize adult hemoglobin in erythroid precursors and in CD34(+) cells isolated from patients affected by β-thalassemia and SCD. Among the thalassemic patients, we identified a subset of specimens in which hemoglobin production can be achieved using fewer copies of the vector integrated than in others. In SCD specimens the treatment with AnkT9W ameliorates erythropoiesis by increasing adult hemoglobin (Hb A) and concurrently reducing the sickling tetramer (Hb S).Our results suggest two major findings. First, we discovered that for the purpose of expressing the β-globin gene the ankyrin element is particularly suitable. Second, our analysis of a large group of specimens from β-thalassemic and SCD patients indicates that clinical trials could benefit from a simple test to predict the relationship between the number of vector copies integrated and the total amount of hemoglobin produced in the erythroid cells of prospective patients. This approach would provide vital information to select the best candidates for these clinical trials, before patients undergo myeloablation and bone marrow transplant.
- Published
- 2012
- Full Text
- View/download PDF
165. β-thalassemia: a model for elucidating the dynamic regulation of ineffective erythropoiesis and iron metabolism.
- Author
-
Ginzburg Y and Rivella S
- Subjects
- Animals, Antimicrobial Cationic Peptides metabolism, Disease Models, Animal, Erythrocytes pathology, Hepcidins, Humans, beta-Thalassemia pathology, Erythrocytes metabolism, Erythropoiesis, Iron metabolism, beta-Thalassemia metabolism, beta-Thalassemia therapy
- Abstract
β-thalassemia is a disease characterized by anemia and is associated with ineffective erythropoiesis and iron dysregulation resulting in iron overload. The peptide hormone hepcidin regulates iron metabolism, and insufficient hepcidin synthesis is responsible for iron overload in minimally transfused patients with this disease. Understanding the crosstalk between erythropoiesis and iron metabolism is an area of active investigation in which patients with and models of β-thalassemia have provided significant insight. The dependence of erythropoiesis on iron presupposes that iron demand for hemoglobin synthesis is involved in the regulation of iron metabolism. Major advances have been made in understanding iron availability for erythropoiesis and its dysregulation in β-thalassemia. In this review, we describe the clinical characteristics and current therapeutic standard in β-thalassemia, explore the definition of ineffective erythropoiesis, and discuss its role in hepcidin regulation. In preclinical experiments using interventions such as transferrin, hepcidin agonists, and JAK2 inhibitors, we provide evidence of potential new treatment alternatives that elucidate mechanisms by which expanded or ineffective erythropoiesis may regulate iron supply, distribution, and utilization in diseases such as β-thalassemia.
- Published
- 2011
- Full Text
- View/download PDF
166. Prospects for a hepcidin mimic to treat β-thalassemia and hemochromatosis.
- Author
-
Parrow NL, Gardenghi S, and Rivella S
- Subjects
- Animals, Antimicrobial Cationic Peptides agonists, Antimicrobial Cationic Peptides therapeutic use, Cation Transport Proteins metabolism, Disease Models, Animal, Hepcidins, Iron metabolism, Mice, Mice, Knockout, Reactive Oxygen Species metabolism, Antimicrobial Cationic Peptides metabolism, Hemochromatosis therapy, beta-Thalassemia therapy
- Published
- 2011
- Full Text
- View/download PDF
167. Enhanced erythropoiesis in Hfe-KO mice indicates a role for Hfe in the modulation of erythroid iron homeostasis.
- Author
-
Ramos P, Guy E, Chen N, Proenca CC, Gardenghi S, Casu C, Follenzi A, Van Rooijen N, Grady RW, de Sousa M, and Rivella S
- Subjects
- Aging blood, Aging metabolism, Aging physiology, Animals, Animals, Newborn, Antimicrobial Cationic Peptides genetics, Antimicrobial Cationic Peptides metabolism, Hemochromatosis Protein, Hepcidins, Histocompatibility Antigens Class I metabolism, Histocompatibility Antigens Class I physiology, Homeostasis genetics, Homeostasis physiology, Iron Overload genetics, Iron Overload metabolism, Membrane Proteins metabolism, Membrane Proteins physiology, Mice, Mice, Inbred C57BL, Mice, Knockout, Stress, Physiological genetics, Stress, Physiological physiology, Transferrin metabolism, Up-Regulation genetics, Up-Regulation physiology, Erythroid Cells metabolism, Erythropoiesis genetics, Histocompatibility Antigens Class I genetics, Iron metabolism, Membrane Proteins genetics
- Abstract
In hereditary hemochromatosis, mutations in HFE lead to iron overload through abnormally low levels of hepcidin. In addition, HFE potentially modulates cellular iron uptake by interacting with transferrin receptor, a crucial protein during erythropoiesis. However, the role of HFE in this process was never explored. We hypothesize that HFE modulates erythropoiesis by affecting dietary iron absorption and erythroid iron intake. To investigate this, we used Hfe-KO mice in conditions of altered dietary iron and erythropoiesis. We show that Hfe-KO mice can overcome phlebotomy-induced anemia more rapidly than wild-type mice (even when iron loaded). Second, we evaluated mice combining the hemochromatosis and β-thalassemia phenotypes. Our results suggest that lack of Hfe is advantageous in conditions of increased erythropoietic activity because of augmented iron mobilization driven by deficient hepcidin response. Lastly, we demonstrate that Hfe is expressed in erythroid cells and impairs iron uptake, whereas its absence exclusively from the hematopoietic compartment is sufficient to accelerate recovery from phlebotomy. In summary, we demonstrate that Hfe influences erythropoiesis by 2 distinct mechanisms: limiting hepcidin expression under conditions of simultaneous iron overload and stress erythropoiesis, and impairing transferrin-bound iron uptake by erythroid cells. Moreover, our results provide novel suggestions to improve the treatment of hemochromatosis.
- Published
- 2011
- Full Text
- View/download PDF
168. Anemia, ineffective erythropoiesis, and hepcidin: interacting factors in abnormal iron metabolism leading to iron overload in β-thalassemia.
- Author
-
Gardenghi S, Grady RW, and Rivella S
- Subjects
- Animals, Erythropoietin metabolism, Hepcidins, Humans, Iron metabolism, Iron Overload, Receptors, Erythropoietin metabolism, Anemia metabolism, Antimicrobial Cationic Peptides metabolism, Erythropoiesis, beta-Thalassemia metabolism
- Abstract
β-Thalassemia is a genetic disorder caused by mutations in the β-globin gene and characterized by chronic anemia caused by ineffective erythropoiesis, and accompanied by a variety of serious secondary complications such as extramedullary hematopoiesis, splenomegaly, and iron overload. In the past few years, numerous studies have shown that such secondary disease conditions have a genetic basis caused by the abnormal expression of genes with a role in controlling erythropoiesis and iron metabolism. In this article, the most recent discoveries related to the mechanism(s) responsible for anemia/ineffective erythropoiesis and iron overload are discussed in detail. Particular attention is paid to the pathway(s) controlling the expression of hepcidin, which is the main regulator of iron metabolism, and the Epo/EpoR/Jak2/Stat5 signaling pathway, which regulates erythropoiesis. Better understanding of how these pathways function and are altered in β-thalassemia has revealed several possibilities for development of new therapeutic approaches to treat of the complications of this disease., (Published by Elsevier Inc.)
- Published
- 2010
- Full Text
- View/download PDF
169. Hepcidin as a therapeutic tool to limit iron overload and improve anemia in β-thalassemic mice.
- Author
-
Gardenghi S, Ramos P, Marongiu MF, Melchiori L, Breda L, Guy E, Muirhead K, Rao N, Roy CN, Andrews NC, Nemeth E, Follenzi A, An X, Mohandas N, Ginzburg Y, Rachmilewitz EA, Giardina PJ, Grady RW, and Rivella S
- Subjects
- Animals, Antimicrobial Cationic Peptides genetics, Base Sequence, DNA Primers genetics, Disease Models, Animal, Erythropoiesis drug effects, Gene Expression, Hepcidins, Humans, Iron metabolism, Iron Overload blood, Iron Overload metabolism, Iron, Dietary administration & dosage, Mice, Mice, Inbred C57BL, Mice, Mutant Strains, Mice, Transgenic, Recombinant Proteins genetics, Recombinant Proteins therapeutic use, beta-Thalassemia blood, beta-Thalassemia metabolism, Antimicrobial Cationic Peptides therapeutic use, Iron Overload drug therapy, beta-Thalassemia drug therapy
- Abstract
Excessive iron absorption is one of the main features of β-thalassemia and can lead to severe morbidity and mortality. Serial analyses of β-thalassemic mice indicate that while hemoglobin levels decrease over time, the concentration of iron in the liver, spleen, and kidneys markedly increases. Iron overload is associated with low levels of hepcidin, a peptide that regulates iron metabolism by triggering degradation of ferroportin, an iron-transport protein localized on absorptive enterocytes as well as hepatocytes and macrophages. Patients with β-thalassemia also have low hepcidin levels. These observations led us to hypothesize that more iron is absorbed in β-thalassemia than is required for erythropoiesis and that increasing the concentration of hepcidin in the body of such patients might be therapeutic, limiting iron overload. Here we demonstrate that a moderate increase in expression of hepcidin in β-thalassemic mice limits iron overload, decreases formation of insoluble membrane-bound globins and reactive oxygen species, and improves anemia. Mice with increased hepcidin expression also demonstrated an increase in the lifespan of their red cells, reversal of ineffective erythropoiesis and splenomegaly, and an increase in total hemoglobin levels. These data led us to suggest that therapeutics that could increase hepcidin levels or act as hepcidin agonists might help treat the abnormal iron absorption in individuals with β-thalassemia and related disorders.
- Published
- 2010
- Full Text
- View/download PDF
170. Analysis of alpha hemoglobin stabilizing protein overexpression in murine β-thalassemia.
- Author
-
Nasimuzzaman M, Khandros E, Wang X, Kong Y, Zhao H, Weiss D, Rivella S, Weiss MJ, and Persons DA
- Subjects
- Amino Acid Substitution, Animals, Blood Proteins deficiency, Blood Proteins genetics, Disease Models, Animal, Erythrocyte Indices, Erythroid Cells metabolism, Genetic Vectors genetics, Genetic Vectors therapeutic use, Hemoglobins analysis, Humans, Mice, Mice, Knockout, Mice, Transgenic, Molecular Chaperones genetics, Radiation Chimera, Reactive Oxygen Species, Recombinant Fusion Proteins physiology, Species Specificity, beta-Thalassemia genetics, beta-Thalassemia physiopathology, Blood Proteins physiology, Molecular Chaperones physiology, alpha-Globins metabolism, beta-Thalassemia blood
- Abstract
Excess free alpha-globin is cytotoxic and contributes to the pathophysiology of b-thalassemia. Alpha hemoglobin stabilizing protein (AHSP) is a molecular chaperone that binds free alpha-globin to promote its folding and inhibit its ability to produce damaging reactive oxygen species. Reduced AHSP levels correlate with increased severity of b-thalassemia in some human cohorts, but causal mechanistic relationships are not established for these associations. We used transgenic and lentiviral gene transfer methods to investigate whether supraphysiologic AHSP levels could mitigate the severity of b-thalassemia intermedia by providing an increased sink for the excess pool of alpha-globin chains. We tested wild-type AHSP and two mutant versions with amino acid substitutions that confer 3- or 13-fold higher affinity for alpha-globin. Erythroid overexpression of these AHSP proteins up to 11-fold beyond endogenous levels had no major effects on hematologic parameters in b-thalassemic animals. Our results demonstrate that endogenous AHSP is not limiting for a-globin detoxification in a murine model of b-thalassemia.
- Published
- 2010
- Full Text
- View/download PDF
171. Hepcidin and Hfe in iron overload in beta-thalassemia.
- Author
-
Gardenghi S, Ramos P, Follenzi A, Rao N, Rachmilewitz EA, Giardina PJ, Grady RW, and Rivella S
- Subjects
- Anemia etiology, Anemia metabolism, Animals, Anti-Bacterial Agents metabolism, Anti-Bacterial Agents therapeutic use, Antimicrobial Cationic Peptides genetics, Antimicrobial Cationic Peptides therapeutic use, Genetic Therapy, Genetic Vectors genetics, Hemochromatosis Protein, Hepcidins, Histocompatibility Antigens Class I genetics, Histocompatibility Antigens Class I therapeutic use, Humans, Iron Overload etiology, Iron Overload therapy, Membrane Proteins genetics, Membrane Proteins therapeutic use, beta-Thalassemia complications, beta-Thalassemia genetics, beta-Thalassemia therapy, Antimicrobial Cationic Peptides metabolism, Histocompatibility Antigens Class I metabolism, Iron Overload metabolism, Membrane Proteins metabolism, beta-Thalassemia metabolism
- Abstract
Hepcidin (HAMP) negatively regulates iron absorption, degrading the iron exporter ferroportin at the level of enterocytes and macrophages. We showed that mice with beta-thalassemia intermedia (th3/+) have increased anemia and iron overload. However, their hepcidin expression is relatively low compared to their iron burden. We also showed that the iron metabolism gene Hfe is down-regulated in concert with hepcidin in th3/+ mice. These observations suggest that low hepcidin levels are responsible for abnormal iron absorption in thalassemic mice and that down-regulation of Hfe might be involved in the pathway that controls hepcidin synthesis in beta-thalassemia. Therefore, these studies suggest that increasing hepcidin and/or Hfe expression could be a strategy to reduces iron overload in these animals. The goal of this paper is to review recent findings that correlate hepcidin, Hfe, and iron metabolism in beta-thalassemia and to discuss potential novel therapeutic approaches based on these recent discoveries.
- Published
- 2010
- Full Text
- View/download PDF
172. Iron metabolism and ineffective erythropoiesis in beta-thalassemia mouse models.
- Author
-
Ramos P, Melchiori L, Gardenghi S, Van-Roijen N, Grady RW, Ginzburg Y, and Rivella S
- Subjects
- Animals, Anti-Bacterial Agents metabolism, Antimicrobial Cationic Peptides metabolism, Hepcidins, Humans, Iron Overload metabolism, Mice, Disease Models, Animal, Erythropoiesis physiology, Iron metabolism, beta-Thalassemia metabolism, beta-Thalassemia physiopathology
- Abstract
beta-thalassemia is a disease associated with decreased beta-globin production leading to anemia, ineffective erythropoiesis, and iron overload. New mechanisms associated with modulation of erythropoiesis and iron metabolism have recently been discovered in thalassemic mice, improving our understanding of the pathophysiology of this disease. These discoveries have the potential to be translated into clinically-relevant therapeutic options to reduce ineffective erythropoiesis and iron overload. A new generation of therapies based on limiting ineffective erythropoiesis, iron absorption, and the correction of iron maldistribution could be on the way, possibly complementing and improving the current standard of patient care.
- Published
- 2010
- Full Text
- View/download PDF
173. A preclinical approach for gene therapy of beta-thalassemia.
- Author
-
Breda L, Kleinert DA, Casu C, Casula L, Cartegni L, Fibach E, Mancini I, Giardina PJ, Gambari R, and Rivella S
- Subjects
- Animals, Cells, Cultured, Genetic Vectors genetics, Genetic Vectors therapeutic use, Humans, Lentivirus genetics, Mice, Mutation, beta-Globins genetics, beta-Globins metabolism, Genetic Therapy, beta-Thalassemia genetics, beta-Thalassemia therapy
- Abstract
Lentiviral-mediated beta-globin gene transfer successfully treated beta-thalassemic mice. Based on this result, clinical trials were initiated. To date, however, no study has investigated the efficacy of gene therapy in relation to the nature of the different beta-globin mutations found in patients. Most mutations can be classified as beta(0) or beta(+), based on the amount of beta-globin protein produced. Therefore, we propose that a screening in vitro is necessary to verify the efficacy of gene transfer prior to treatment of individual patients. We used a two-phase liquid culture system to expand and differentiate erythroid progenitor cells (ErPCs) transduced with lentiviral vectors. We propose the use of this system to test the efficiency of lentiviral vectors carrying the human beta-globin gene, to correct the phenotype of ErPCs from patients preparing for gene therapy. This new approach might have profound implications for designing gene therapy and for understanding the genotype/phenotype variability observed in Cooley's anemia patients.
- Published
- 2010
- Full Text
- View/download PDF
174. Changes in bone microarchitecture and biomechanical properties in the th3 thalassemia mouse are associated with decreased bone turnover and occur during the period of bone accrual.
- Author
-
Vogiatzi MG, Tsay J, Verdelis K, Rivella S, Grady RW, Doty S, Giardina PJ, and Boskey AL
- Subjects
- Animals, Biomechanical Phenomena, Bone and Bones, Erythropoiesis, Mice, Mice, Inbred C57BL, Osteoporosis etiology, Reverse Transcriptase Polymerase Chain Reaction, Spectroscopy, Fourier Transform Infrared, Thalassemia genetics, Tomography, X-Ray Computed, Bone Remodeling physiology, Osteoporosis pathology, Thalassemia complications
- Abstract
Osteoporosis and fractures occur frequently in patients with beta-thalassemias, a group of congenital hemolytic anemias characterized by decreased synthesis of the beta chain of hemoglobin. In this study, we determined the bone abnormalities of the th3 thalassemia mouse, generated by deletion of the mouse beta-chain genes. The heterozygous th3/+ mouse has moderate anemia and serves as a model of beta-thalassemia intermedia, which represents the mild thalassemia phenotype. The th3/th3 mouse has lethal anemia and is a model of beta-thalassemia major, which is characterized by life-threatening anemia requiring regular transfusions to sustain life. Compared to controls, (1) microCT of trabecular bone showed decreased bone volume fraction, number of trabeculae, and trabecular thickness in both th3/+ and th3/th3 (P < 0.05); (2) cortical bone analysis showed thinner cortices and increased marrow area in th3/+ (P < 0.05); (3) microCT abnormalities in th3/+ mice were present by 2 months and did not worsen with age; (4) histomorphometry was significant for decreased bone formation and resorption in both th3/+ and th3/th3, and expression of cathepsin K and osteocalcin from bone of both th3/+ and th3/th3 animals was reduced (P < 0.05); (5) biomechanics showed reduced maximum load, maximum moment, and structural stiffness in both th3/+ and th3/th3 (P < 0.01). In conclusion, the th3 mouse model of thalassemia manifests bone changes reminiscent of those in humans and can be used for further bone studies in thalassemia. Bone changes are associated with decreased bone turnover and develop early during the period of bone accrual.
- Published
- 2010
- Full Text
- View/download PDF
175. Crosstalk between Erythropoiesis and Iron Metabolism.
- Author
-
Rivella S, Nemeth E, and Miller JL
- Published
- 2010
- Full Text
- View/download PDF
176. Future alternative therapies for β-thalassemia.
- Author
-
Rivella S and Rachmilewitz E
- Abstract
β-thalassemia is an inherited disorder due to mutations found in the β-globin gene, leading to anemia and requiring sporadic or chronic blood transfusions for survival. Without proper chelation, β-thalassemia results in iron overload. Ineffective erythropoiesis can lead to iron overload even in untransfused patients who are affected by β-thalassemia intermedia. Better understanding of the molecular biologic aspects of this disorder has led to improvements in population screening and prenatal diagnosis, which, in turn, have led to dramatic reductions in the number of children born with β-thalassemia major in the Mediterranean littoral. However, as a consequence of decreases in neonatal and childhood mortality in other geographical areas, β-thalassemia has become a worldwide clinical problem. A number of unsolved pathophysiological issues remain, such as ineffective erythropoieis, abnormal iron absorption, oxidative stress, splenomegaly and thrombosis. In the last few years, novel studies have the potential to introduce new therapeutic approaches that might reduce these problems and limit the need for blood transfusion.
- Published
- 2009
- Full Text
- View/download PDF
177. Id1 represses osteoclast-dependent transcription and affects bone formation and hematopoiesis.
- Author
-
Chan AS, Jensen KK, Skokos D, Doty S, Lederman HK, Kaplan RN, Rafii S, Rivella S, and Lyden D
- Subjects
- Animals, Bone Marrow Cells cytology, Female, Gene Expression Regulation, Hematopoietic Stem Cells cytology, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Phenotype, Bone and Bones physiology, Inhibitor of Differentiation Protein 1 metabolism, Osteoclasts metabolism, Osteogenesis genetics, Transcription, Genetic
- Abstract
Background: The bone-bone marrow interface is an area of the bone marrow microenvironment in which both bone remodeling cells, osteoblasts and osteoclasts, and hematopoietic cells are anatomically juxtaposed. The close proximity of these cells naturally suggests that they interact with one another, but these interactions are just beginning to be characterized., Methodology/principal Findings: An Id1(-/-) mouse model was used to assess the role of Id1 in the bone marrow microenvironment. Micro-computed tomography and fracture tests showed that Id1(-/-) mice have reduced bone mass and increased bone fragility, consistent with an osteoporotic phenotype. Osteoclastogenesis and pit formation assays revealed that loss of Id1 increased osteoclast differentiation and resorption activity, both in vivo and in vitro, suggesting a cell autonomous role for Id1 as a negative regulator of osteoclast differentiation. Examination by flow cytometry of the hematopoietic compartment of Id1(-/-) mice showed an increase in myeloid differentiation. Additionally, we found increased expression of osteoclast genes, TRAP, Oscar, and CTSK in the Id1(-/-) bone marrow microenvironment. Lastly, transplantation of wild-type bone marrow into Id1(-/-) mice repressed TRAP, Oscar, and CTSK expression and activity and rescued the hematopoietic and bone phenotype in these mice., Conclusions/significance: In conclusion, we demonstrate an osteoporotic phenotype in Id1(-/-) mice and a mechanism for Id1 transcriptional control of osteoclast-associated genes. Our results identify Id1 as a principal player responsible for the dynamic cross-talk between bone and bone marrow hematopoietic cells.
- Published
- 2009
- Full Text
- View/download PDF
178. Gene therapy in thalassemia and hemoglobinopathies.
- Author
-
Breda L, Gambari R, and Rivella S
- Abstract
Sickle cell disease (SCD) and ß-thalassemia represent the most common hemoglobinopathies caused, respectively, by the alteration of structural features or deficient production of the ß-chain of the Hb molecule. Other hemoglobinopathies are characterized by different mutations in the α- or ß-globin genes and are associated with anemia and might require periodic or chronic blood transfusions. Therefore, ß-thalassemia, SCD and other hemoglobinopathies are excellent candidates for genetic approaches since they are monogenic disorders and, potentially, could be cured by introducing or correcting a single gene into the hematopoietic compartment or a single stem cell. Initial attempts at gene transfer of these hemoglobinopathies have proved unsuccessful due to limitations of available gene transfer vectors. With the advent of lentiviral vectors many of the initial limitations have been overcame. New approaches have also focused on targeting the specific mutation in the ß-globin genes, correcting the DNA sequence or manipulating the fate of RNA translation and splicing to restore ß-globin chain synthesis. These techniques have the potential to correct the defect into hematopoietic stem cells or be utilized to modify stem cells generated from patients affected by these disorders. This review discusses gene therapy strategies for the hemoglobinopathies, including the use of lentiviral vectors, generation of induced pluripotent stem cells (iPS) cells, gene targeting, splice-switching and stop codon readthrough.
- Published
- 2009
- Full Text
- View/download PDF
179. Production of beta-globin and adult hemoglobin following G418 treatment of erythroid precursor cells from homozygous beta(0)39 thalassemia patients.
- Author
-
Salvatori F, Breveglieri G, Zuccato C, Finotti A, Bianchi N, Borgatti M, Feriotto G, Destro F, Canella A, Brognara E, Lampronti I, Breda L, Rivella S, and Gambari R
- Subjects
- Cells, Cultured, Erythroid Precursor Cells cytology, Hemoglobins genetics, Homozygote, Humans, K562 Cells, beta-Globins drug effects, beta-Globins genetics, beta-Thalassemia blood, beta-Thalassemia genetics, Codon, Nonsense, Erythroid Precursor Cells metabolism, Gentamicins pharmacology, Hemoglobins biosynthesis, beta-Globins biosynthesis, beta-Thalassemia drug therapy
- Abstract
In several types of thalassemia (including beta(0)39-thalassemia), stop codon mutations lead to premature translation termination and to mRNA destabilization through nonsense-mediated decay. Drugs (for instance aminoglycosides) can be designed to suppress premature termination, inducing a ribosomal readthrough. These findings have introduced new hopes for the development of a pharmacologic approach to the cure of this disease. However, the effects of aminoglycosides on globin mRNA carrying beta-thalassemia stop mutations have not yet been investigated. In this study, we have used a lentiviral construct containing the beta(0)39-thalassemia globin gene under control of the beta-globin promoter and a LCR cassette. We demonstrated by fluorescence-activated cell sorting (FACS) analysis the production of beta-globin by K562 cell clones expressing the beta(0)39-thalassemia globin gene and treated with G418. More importantly, after FACS and high-performance liquid chromatography (HPLC) analyses, erythroid precursor cells from beta(0)39-thalassemia patients were demonstrated to be able to produce beta-globin and adult hemoglobin after treatment with G418. This study strongly suggests that ribosomal readthrough should be considered a strategy for developing experimental strategies for the treatment of beta(0)-thalassemia caused by stop codon mutations. Am. J. Hematol., 2009. (c) 2009 Wiley-Liss, Inc.
- Published
- 2009
- Full Text
- View/download PDF
180. Unexpected expression of alpha- and beta-globin in mesencephalic dopaminergic neurons and glial cells.
- Author
-
Biagioli M, Pinto M, Cesselli D, Zaninello M, Lazarevic D, Roncaglia P, Simone R, Vlachouli C, Plessy C, Bertin N, Beltrami A, Kobayashi K, Gallo V, Santoro C, Ferrer I, Rivella S, Beltrami CA, Carninci P, Raviola E, and Gustincich S
- Subjects
- Animals, Flow Cytometry, Green Fluorescent Proteins, Humans, Mice, Oligonucleotide Array Sequence Analysis, Rats, Neuroglia metabolism, Neurons metabolism, Substantia Nigra cytology, Ventral Tegmental Area cytology, alpha-Globins metabolism, beta-Globins metabolism
- Abstract
The mesencephalic dopaminergic (mDA) cell system is composed of two major groups of projecting cells in the substantia nigra (SN) (A9 neurons) and the ventral tegmental area (VTA) (A10 cells). A9 neurons form the nigrostriatal pathway and are involved in regulating voluntary movements and postural reflexes. Their selective degeneration leads to Parkinson's disease. Here, we report that gene expression analysis of A9 dopaminergic neurons (DA) identifies transcripts for alpha- and beta-chains of hemoglobin (Hb). Globin immunoreactivity decorates the majority of A9 DA, a subpopulation of cortical and hippocampal astrocytes and mature oligodendrocytes. This pattern of expression was confirmed in different mouse strains and in rat and human. We show that Hb is expressed in the SN of human postmortem brain. By microarray analysis of dopaminergic cell lines overexpressing alpha- and beta-globin chains, changes in genes involved in O(2) homeostasis and oxidative phopshorylation were observed, linking Hb expression to mitochondrial function. Our data suggest that the most famed oxygen-carrying globin is not exclusively restricted to the blood, but it may play a role in the normal physiology of the brain and neurodegenerative diseases.
- Published
- 2009
- Full Text
- View/download PDF
181. Development of K562 cell clones expressing beta-globin mRNA carrying the beta039 thalassaemia mutation for the screening of correctors of stop-codon mutations.
- Author
-
Salvatori F, Cantale V, Breveglieri G, Zuccato C, Finotti A, Bianchi N, Borgatti M, Feriotto G, Destro F, Canella A, Breda L, Rivella S, and Gambari R
- Subjects
- Clone Cells, Cloning, Molecular, Gene Expression drug effects, Gentamicins pharmacology, Green Fluorescent Proteins biosynthesis, Green Fluorescent Proteins genetics, Humans, Lentivirus genetics, Mutagenesis, Site-Directed, Polymerase Chain Reaction, RNA, Messenger genetics, Recombinant Fusion Proteins biosynthesis, Recombinant Fusion Proteins genetics, Transcription, Genetic drug effects, Transfection, beta-Globins biosynthesis, Codon, Nonsense, K562 Cells physiology, Point Mutation, RNA, Messenger biosynthesis, beta-Globins genetics, beta-Thalassemia genetics
- Abstract
Nonsense mutations, giving rise to UAA, UGA and UAG stop codons within the coding region of mRNAs, promote premature translational termination and are the leading cause of approx. 30% of inherited diseases, including cystic fibrosis, Duchenne muscular dystrophy and thalassaemia. For instance, in beta(0)39-thalassaemia the CAG (glutamine) codon is mutated to the UAG stop codon, leading to premature translation termination and to mRNA destabilization through the well-described NMD (nonsense-mediated mRNA decay). In order to develop an approach facilitating translation and, therefore, protection from NMD, aminoglycoside antibiotics have been tested on mRNAs carrying premature stop codons. These drugs decrease the accuracy in the codon-anticodon base-pairing, inducing a ribosomal read-through of the premature termination codons. Interestingly, recent papers have described drugs designed and produced for suppressing premature translational termination, inducing a ribosomal read-through of premature but not normal termination codons. These findings have introduced new hopes for the development of a pharmacological approach to the therapy of beta(0)39-thalassaemia. In this context, we started the development of a cellular model of the beta(0)39-thalassaemia mutation that could be used for the screening of a high number of aminoglycosides and analogous molecules. To this aim, we produced a lentiviral construct containing the beta(0)39-thalassaemia globin gene under a minimal LCR (locus control region) control and used this construct for the transduction of K562 cells, subsequently subcloned, with the purpose to obtain several K562 clones with different integration copies of the construct. These clones were then treated with Geneticin (also known as G418) and other aminoglycosides and the production of beta-globin was analysed by FACS analysis. The results obtained suggest that this experimental system is suitable for the characterization of correction of the beta(0)39-globin mutation causing beta-thalassaemia.
- Published
- 2009
- Full Text
- View/download PDF
182. Regulation of iron absorption in hemoglobinopathies.
- Author
-
Rechavi G and Rivella S
- Subjects
- Animals, Disease Models, Animal, Globins genetics, Hemoglobinopathies complications, Humans, Iron Overload complications, Hemoglobinopathies metabolism, Intestinal Absorption, Iron metabolism
- Abstract
Beta-thalassemia and sickle cell anemia (SCD) represent the most common hemoglobinopathies caused, respectively, by deficient production or alteration of the beta chain of hemoglobin (Hb). Patients affected by the most severe form of thalassemia suffer from profound anemia that requires chronic blood transfusions and chelation therapies to prevent iron overload. However, patients affected by beta-thalassemia intermedia, a milder form of the disease that does not require chronic blood transfusions, eventually also show elevated body iron content due to increased gastrointestinal iron absorption. Even SCD patients might require blood transfusions and iron chelation to prevent deleterious and painful vaso-occlusive crises and complications due to iron overload. Although definitive cures are presently available, such as bone marrow transplantation (BMT), or are in development, such as correction of the disease through hematopoietic stem cell beta-globin gene transfer, they are potentially hazardous procedures or too experimental to provide consistently safe and predictive clinical outcomes. Therefore, studies that aim to better understand the pathophysiology of the hemoglobinopathies might provide further insight and new drugs to dramatically improve the understanding and current treatment of these diseases. This review will describe how recent discoveries on iron metabolism and erythropoiesis could lead to new therapeutic strategies and better clinical care of these diseases, thereby yielding a much better quality of life for the patients.
- Published
- 2008
- Full Text
- View/download PDF
183. Decreased differentiation of erythroid cells exacerbates ineffective erythropoiesis in beta-thalassemia.
- Author
-
Libani IV, Guy EC, Melchiori L, Schiro R, Ramos P, Breda L, Scholzen T, Chadburn A, Liu Y, Kernbach M, Baron-Lühr B, Porotto M, de Sousa M, Rachmilewitz EA, Hood JD, Cappellini MD, Giardina PJ, Grady RW, Gerdes J, and Rivella S
- Subjects
- Animals, Apoptosis, Cyclin-Dependent Kinases genetics, Janus Kinase 2 antagonists & inhibitors, Mice, Spleen pathology, Cell Differentiation, Erythroid Cells pathology, Erythropoiesis, Janus Kinase 2 genetics, beta-Thalassemia blood
- Abstract
In beta-thalassemia, the mechanism driving ineffective erythropoiesis (IE) is insufficiently understood. We analyzed mice affected by beta-thalassemia and observed, unexpectedly, a relatively small increase in apoptosis of their erythroid cells compared with healthy mice. Therefore, we sought to determine whether IE could also be characterized by limited erythroid cell differentiation. In thalassemic mice, we observed that a greater than normal percentage of erythroid cells was in S-phase, exhibiting an erythroblast-like morphology. Thalassemic cells were associated with expression of cell cycle-promoting genes such as EpoR, Jak2, Cyclin-A, Cdk2, and Ki-67 and the antiapoptotic protein Bcl-X(L). The cells also differentiated less than normal erythroid ones in vitro. To investigate whether Jak2 could be responsible for the limited cell differentiation, we administered a Jak2 inhibitor, TG101209, to healthy and thalassemic mice. Exposure to TG101209 dramatically decreased the spleen size but also affected anemia. Although our data do not exclude a role for apoptosis in IE, we propose that expansion of the erythroid pool followed by limited cell differentiation exacerbates IE in thalassemia. In addition, these results suggest that use of Jak2 inhibitors has the potential to profoundly change the management of this disorder.
- Published
- 2008
- Full Text
- View/download PDF
184. Protective role of calreticulin in HFE hemochromatosis.
- Author
-
Pinto JP, Ramos P, de Almeida SF, Oliveira S, Breda L, Michalak M, Porto G, Rivella S, and de Sousa M
- Subjects
- Adult, Aged, Animals, Antimicrobial Cationic Peptides genetics, Antimicrobial Cationic Peptides metabolism, Calreticulin antagonists & inhibitors, Cell Line, Disease Models, Animal, Female, Gene Expression Regulation, Hemochromatosis physiopathology, Hemochromatosis Protein, Hepatocytes metabolism, Hepatocytes pathology, Hepcidins, Humans, Male, Mice, Middle Aged, Monocytes metabolism, Monocytes pathology, Oxidative Stress, RNA, Messenger analysis, RNA, Small Interfering administration & dosage, Rats, Receptors, Transferrin biosynthesis, Receptors, Transferrin blood, Receptors, Transferrin genetics, Transfection, Calreticulin blood, Calreticulin genetics, Hemochromatosis genetics, Histocompatibility Antigens Class I genetics, Histocompatibility Antigens Class I metabolism, Membrane Proteins genetics, Membrane Proteins metabolism, Mutation, Missense
- Abstract
HFE gene mutations are associated with over 80% of cases of hereditary hemochromatosis (HH), an iron-overload disease in which the liver is the most frequently affected organ. Research on HFE has traditionally focused on its interaction with the transferrin receptor. More recent studies have suggested a more complex function for this nonclassical MHC-I protein. The aim of this study was to examine how HFE and its two most common mutations affect the expression of selected genes in a hepatocyte-like cell line. Gene expression was analyzed in HepG2 cells overexpressing wild-type and mutant HFE. The effect of HFE in iron import and oxidative stress levels was assessed. Unfolded protein response (UPR)-activated gene expression was analyzed in peripheral blood mononuclear cells from characterized HH patients. C282Y HFE down-regulated hepcidin and enhanced calreticulin mRNA expression. Calreticulin levels correlated with intracellular iron increase and were associated with protection from oxidative stress. In C282Y(+/+) patients calreticulin levels correlated with the expression of the UPR marker BiP and showed a negative association with the number of hereditary hemochromatosis clinical manifestations. The data show that expression of C282Y HFE triggers a stress-protective response in HepG2 cells and suggest a role for calreticulin as a modifier of the clinical expression of HH.
- Published
- 2008
- Full Text
- View/download PDF
185. Ineffective erythropoiesis in beta-thalassemia is characterized by increased iron absorption mediated by down-regulation of hepcidin and up-regulation of ferroportin.
- Author
-
Gardenghi S, Marongiu MF, Ramos P, Guy E, Breda L, Chadburn A, Liu Y, Amariglio N, Rechavi G, Rachmilewitz EA, Breuer W, Cabantchik ZI, Wrighting DM, Andrews NC, de Sousa M, Giardina PJ, Grady RW, and Rivella S
- Subjects
- Animals, Blood Transfusion, Flow Cytometry, Hepcidins, Iron metabolism, Mice, Mice, Inbred C57BL, Mice, Transgenic, Phenotype, beta-Thalassemia metabolism, Antimicrobial Cationic Peptides biosynthesis, Cation Transport Proteins biosynthesis, Down-Regulation, Erythropoiesis, Gene Expression Regulation, Iron pharmacokinetics, Up-Regulation, beta-Thalassemia blood
- Abstract
Progressive iron overload is the most salient and ultimately fatal complication of beta-thalassemia. However, little is known about the relationship among ineffective erythropoiesis (IE), the role of iron-regulatory genes, and tissue iron distribution in beta-thalassemia. We analyzed tissue iron content and iron-regulatory gene expression in the liver, duodenum, spleen, bone marrow, kidney, and heart of mice up to 1 year old that exhibit levels of iron overload and anemia consistent with both beta-thalassemia intermedia (th3/+) and major (th3/th3). Here we show, for the first time, that tissue and cellular iron distribution are abnormal and different in th3/+ and th3/th3 mice, and that transfusion therapy can rescue mice affected by beta-thalassemia major and modify both the absorption and distribution of iron. Our study reveals that the degree of IE dictates tissue iron distribution and that IE and iron content regulate hepcidin (Hamp1) and other iron-regulatory genes such as Hfe and Cebpa. In young th3/+ and th3/th3 mice, low Hamp1 levels are responsible for increased iron absorption. However, in 1-year-old th3/+ animals, Hamp1 levels rise and it is rather the increase of ferroportin (Fpn1) that sustains iron accumulation, thus revealing a fundamental role of this iron transporter in the iron overload of beta-thalassemia.
- Published
- 2007
- Full Text
- View/download PDF
186. Therapeutic options for patients with severe beta-thalassemia: the need for globin gene therapy.
- Author
-
Sadelain M, Boulad F, Galanello R, Giardina P, Locatelli F, Maggio A, Rivella S, Riviere I, and Tisdale J
- Subjects
- Animals, Humans, Mice, beta-Thalassemia genetics, beta-Thalassemia pathology, Genetic Therapy methods, Globins genetics, beta-Thalassemia therapy
- Published
- 2007
- Full Text
- View/download PDF
187. Downregulation of hepcidin and haemojuvelin expression in the hepatocyte cell-line HepG2 induced by thalassaemic sera.
- Author
-
Weizer-Stern O, Adamsky K, Amariglio N, Levin C, Koren A, Breuer W, Rachmilewitz E, Breda L, Rivella S, Cabantchik ZI, and Rechavi G
- Subjects
- Acute-Phase Proteins biosynthesis, Acute-Phase Proteins genetics, Antimicrobial Cationic Peptides genetics, Blood Transfusion, Cell Line, GPI-Linked Proteins, Hemochromatosis blood, Hemochromatosis Protein, Hepcidins, Humans, Lipocalin-2, Lipocalins, Membrane Proteins genetics, Polymerase Chain Reaction methods, Proto-Oncogene Proteins biosynthesis, Proto-Oncogene Proteins genetics, beta-Thalassemia therapy, Antimicrobial Cationic Peptides biosynthesis, Down-Regulation, Hepatocytes metabolism, Membrane Proteins biosynthesis, beta-Thalassemia blood
- Abstract
Beta-thalassaemia represents a group of diseases, in which ineffective erythropoiesis is accompanied by iron overload. In a mouse model of beta-thalassaemia, we observed that the liver expressed relatively low levels of hepcidin, which is a key factor in the regulation of iron absorption by the gut and of iron recycling by the reticuloendothelial system. It was hypothesised that, despite the overt iron overload, a putative plasma factor found in beta-thalassaemia might suppress liver hepcidin expression. Sera from beta-thalassaemia and haemochromatosis (C282Y mutation) patients were compared with those of healthy individuals regarding their capacity to induce changes the expression of key genes of iron metabolism in human HepG2 hepatoma cells. Sera from beta-thalassaemia major patients induced a major decrease in hepcidin (HAMP) and lipocalin2 (oncogene 24p3) (LCN2) expression, as well as a moderate decrease in haemojuvelin (HFE2) expression, compared with sera from healthy individuals. A significant correlation was found between the degree of downregulation of HAMP and HFE2 induced by beta-thalassaemia major sera (r = 0.852, P < 0.0009). Decreased HAMP expression was also found in HepG2 cells treated with sera from beta-thalassaemia intermedia patients. In contrast, the majority of sera from hereditary haemochromatosis patients induced an increase in HAMP expression, which correlated with transferrin (Tf) saturation (r = 0.765, P < 0.0099). Our results suggest that, in beta-thalassaemia, serum factors might override the potential effect of iron overload on HAMP expression, thereby providing an explanation for the failure to arrest excessive intestinal iron absorption in these patients.
- Published
- 2006
- Full Text
- View/download PDF
188. mRNA expression of iron regulatory genes in beta-thalassemia intermedia and beta-thalassemia major mouse models.
- Author
-
Weizer-Stern O, Adamsky K, Amariglio N, Rachmilewitz E, Breda L, Rivella S, and Rechavi G
- Subjects
- Acute-Phase Proteins biosynthesis, Acute-Phase Proteins genetics, Animals, Antimicrobial Cationic Peptides urine, Disease Models, Animal, Down-Regulation genetics, Erythroid Precursor Cells metabolism, Erythroid Precursor Cells pathology, Erythropoiesis genetics, Female, Gene Expression Profiling, Globins genetics, Globins metabolism, Hepcidins, Humans, Iron Overload genetics, Iron Overload metabolism, Lipocalin-2, Lipocalins, Mice, Oncogene Proteins biosynthesis, Oncogene Proteins genetics, Receptors, Transferrin biosynthesis, Receptors, Transferrin genetics, beta-Thalassemia metabolism, beta-Thalassemia pathology, Antimicrobial Cationic Peptides genetics, Genes, Regulator genetics, Intestinal Absorption genetics, Iron metabolism, beta-Thalassemia genetics
- Abstract
beta-Thalassemia is an inherited anemia in which synthesis of the hemoglobin beta-chain is decreased. The excess unmatched alpha-globin chains accumulate in the growing erythroid precursors, causing their premature death (ineffective erythropoiesis). Clinical features of beta-thalassemia include variably severe anemia and iron accumulation due to increased intestinal iron absorption. The most anemic patients require regular blood transfusions, which exacerbate their iron overload and result in damage to vital organs. The hepatic peptide hepcidin, a key regulator of iron metabolism in mammals, was recently found to be low in the urine of beta-thalassemia patients, compared with healthy controls, despite their iron overload. In our work, we measured by RQ-PCR the liver mRNA expression of hepcidin and other iron regulatory genes in beta-thalassemia major mouse model (C57Bl/6 Hbb(th3/th3)), and compared it with beta-thalassemia intermedia mouse model (C57Bl/6 Hbb(th3/+)) and control mice. We found decreased expression of hepcidin and TfR2 and increased expression of TfR1 and NGAL in the beta-thalassemia mouse models, compared with the control mice. Significant down-regulation of hepcidin expression in beta-thalassemia major, despite iron overload, might explain the increased iron absorption typically observed in thalassemia.
- Published
- 2006
- Full Text
- View/download PDF
189. Role of iron in inducing oxidative stress in thalassemia: Can it be prevented by inhibition of absorption and by antioxidants?
- Author
-
Rachmilewitz EA, Weizer-Stern O, Adamsky K, Amariglio N, Rechavi G, Breda L, Rivella S, and Cabantchik ZI
- Subjects
- Acetylcysteine administration & dosage, Acetylcysteine therapeutic use, Animals, Antimicrobial Cationic Peptides biosynthesis, Antimicrobial Cationic Peptides genetics, Antioxidants administration & dosage, Antioxidants pharmacology, Bone Marrow metabolism, Carcinoma, Hepatocellular pathology, Cell Line, Tumor, Disease Models, Animal, Drug Therapy, Combination, Erythrocytes chemistry, Gene Expression Regulation, Genetic Therapy, Hepcidins, Humans, Intestinal Absorption physiology, Iron adverse effects, Iron blood, Iron chemistry, Iron pharmacokinetics, Iron Chelating Agents administration & dosage, Iron Chelating Agents therapeutic use, Iron, Dietary pharmacokinetics, Liver Neoplasms pathology, Mice, Oxidants chemistry, Oxidants pharmacokinetics, Oxidative Stress, Thalassemia drug therapy, Thalassemia physiopathology, Vitamin E administration & dosage, Vitamin E therapeutic use, Antioxidants therapeutic use, Iron physiology, Thalassemia metabolism
- Abstract
The pathophysiology of thalassemia is, to a certain extent, associated with the generation of labile iron in the pathological red blood cell (RBC). The appearance of such forms of iron at the inner and outer cell surfaces exposes the cell to conditions whereby the labile metal promotes the formation of reactive oxygen species (ROS) leading to cumulative cell damage. Another source of iron accumulation results from increased absorption due to decreased expression of hepcidin. The presence of labile plasma iron (LPI) was carried out using fluorescent probes in the FACS. RNA expression of hepcidin was measured in two models of thalassemic mice. Hepcidin expression was also measured in human hepatoma HepG2 cells following incubation with thalassemic sera. LPI was identified and could be quantitatively measured and correlated with other parameters of iron overload. Hepcidin expression was downregulated in the livers of thalassemic mice, in major more than in intermedia. Thalassemic sera down regulated hepcidin expression in HepG2 liver cells. A possible way to decrease iron absorption could be by modulating hepcidin expression pharmacologically, by gene therapy or by its administration. Treatment with combination of antioxidants such as N-acetylcysteine for proteins and vitamin E for lipids in addition to iron chelators could neutralize the deleterious effects of ROS and monitored by quantitation of LPI.
- Published
- 2005
- Full Text
- View/download PDF
190. Progress toward the genetic treatment of the beta-thalassemias.
- Author
-
Sadelain M, Lisowski L, Samakoglu S, Rivella S, May C, and Riviere I
- Subjects
- Adaptor Proteins, Signal Transducing, Animals, DNA-Binding Proteins genetics, Disease Models, Animal, Gene Expression Regulation, Viral, Gene Silencing, Gene Transfer Techniques, Genetic Vectors adverse effects, Genetic Vectors therapeutic use, Globins biosynthesis, HIV-1 genetics, Hematopoietic Stem Cell Transplantation, Humans, LIM Domain Proteins, Lentivirus genetics, Leukemia, Lymphoid etiology, Locus Control Region genetics, Metalloproteins genetics, Mice, Mice, Inbred C57BL, Mice, Mutant Strains, Mice, Transgenic, Mutagenesis, Insertional, Oncogenes, Proto-Oncogene Proteins, Retroviridae genetics, Severe Combined Immunodeficiency genetics, Severe Combined Immunodeficiency therapy, Terminal Repeat Sequences, Transgenes, Genetic Therapy adverse effects, Globins genetics, beta-Thalassemia therapy
- Abstract
The beta-thalassemias are congenital anemias that are caused by mutations that reduce or abolish expression of the beta-globin gene. They can be cured by allogeneic hematopoietic stem cell (HSC) transplantation, but this therapeutic option is not available to most patients. The transfer of a regulated beta-globin gene in autologous HSCs is a highly attractive alternative treatment. This strategy, which is simple in principle, raises major challenges in terms of controlling expression of the globin transgene, which ideally should be erythroid specific, differentiation- and stage-restricted, elevated, position independent, and sustained over time. Using lentiviral vectors, May et al. demonstrated in 2000 that an optimized combination of proximal and distal transcriptional control elements permits lineage-specific and elevated beta-globin expression, resulting in therapeutic hemoglobin production and correction of anemia in beta-thalassemic mice. Several groups have by now replicated and extended these findings to various mouse models of severe hemoglobinopathies, thus fueling enthusiasm for a potential treatment of beta-thalassemia based on globin gene transfer. Current investigation focuses on safety issues and the need for improved vector production methodologies. The safe implementation of stem cell-based gene therapy requires the prevention of the formation of replication-competent viral genomes and minimization of the risk of insertional oncogenesis. Importantly, globin vectors, in which transcriptional activity is highly restricted, have a lesser risk of activating oncogenes in hematopoietic progenitors than non-tissue-specific vectors, by virtue of their late-stage erythroid specificity. As such, they provide a general paradigm for improving vector safety in stem cell-based gene therapy.
- Published
- 2005
- Full Text
- View/download PDF
191. Exploring the role of hepcidin, an antimicrobial and iron regulatory peptide, in increased iron absorption in beta-thalassemia.
- Author
-
Breda L, Gardenghi S, Guy E, Rachmilewitz EA, Weizer-Stern O, Adamsky K, Amariglio N, Rechavi G, Giardina PJ, Grady RW, and Rivella S
- Subjects
- Animals, Antimicrobial Cationic Peptides biosynthesis, Antimicrobial Cationic Peptides genetics, Carcinoma, Hepatocellular pathology, Cell Line metabolism, Cell Line, Tumor, Disease Models, Animal, Fibroblasts metabolism, Gene Expression Regulation, Genetic Vectors therapeutic use, Hemochromatosis metabolism, Hepatocytes metabolism, Hepcidins, Humans, Iron Overload genetics, Iron Overload metabolism, Lentivirus genetics, Liver metabolism, Liver Neoplasms pathology, Mice, Mice, Knockout, NIH 3T3 Cells, Transduction, Genetic, beta-Thalassemia metabolism, beta-Thalassemia therapy, Antimicrobial Cationic Peptides physiology, Hemochromatosis genetics, Intestinal Absorption physiology, Iron pharmacokinetics, Iron Overload etiology
- Abstract
To develop new treatments for beta-thalassemia, it is essential to identify the genes involved in the relevant pathophysiological processes. Iron metabolism in thalassemia mice being investigated, focusing on the expression of a gene called hepcidin (Hamp), which is expressed in the liver and whose product (Hamp) is secreted into the bloodstream. In mice, iron overload leads to overexpression of Hamp, while Hamp-knockout mice suffer from hemochromatosis. The aim of this study is to investigate Hamp in the mouse model of beta-thalassemia and to address the potential gene transfer of Hamp to prevent abnormal iron absorption.
- Published
- 2005
- Full Text
- View/download PDF
192. Globin gene transfer for treatment of the beta-thalassemias and sickle cell disease.
- Author
-
Sadelain M, Rivella S, Lisowski L, Samakoglu S, and Rivière I
- Subjects
- Anemia, Sickle Cell therapy, Animals, Disease Models, Animal, Globins genetics, Humans, Mice, beta-Thalassemia therapy, Genetic Therapy methods, Globins therapeutic use, Hemoglobinopathies therapy
- Abstract
The beta-thalassemias and sickle cell disease are severe congenital anemias that are caused by mutations that alter the production of the beta chain of hemoglobin. Allogeneic hematopoietic stem cell (HSC) transplantation is curative, but this therapeutic option is not available to the majority of patients. The transfer of a functional globin gene in autologous HCSs thus represents a highly attractive alternative treatment. This strategy, simple in principle, raises major challenges in terms of controlling the expression of the globin transgene, which ideally should be erythroid specific, differentiation-stage restricted, elevated, position independent, and sustained over time. Using lentiviral vectors, we have demonstrated that an optimised combination of proximal and distal transcriptional control elements permits lineage-specific, elevated expression of the beta-globin gene, resulting in therapeutic hemoglobin production and correction of anemia in beta-thalassemic mice. Several groups have now confirmed and extended these findings in various mouse models of severe hemoglobinopathies, thus generating enthusiasm for a genetic treatment based on globin gene transfer. Furthermore, globin vectors represent a general paradigm for the regulation of transgene function and the improvement of vector safety by restricting transgene expression to the differentiated progeny within a single lineage, thereby reducing the risk of activating oncogenes in hematopoietic progenitors. Here we review the principles underlying the genesis of regulated vectors for stem cell therapy.
- Published
- 2004
- Full Text
- View/download PDF
193. Decreased hepcidin mRNA expression in thalassemic mice.
- Author
-
Adamsky K, Weizer O, Amariglio N, Breda L, Harmelin A, Rivella S, Rachmilewitz E, and Rechavi G
- Subjects
- Animals, Hepcidins, Mice, Mice, Inbred C57BL, Antimicrobial Cationic Peptides metabolism, RNA, Messenger metabolism, Thalassemia metabolism
- Published
- 2004
- Full Text
- View/download PDF
194. A novel murine model of Cooley anemia and its rescue by lentiviral-mediated human beta-globin gene transfer.
- Author
-
Rivella S, May C, Chadburn A, Rivière I, and Sadelain M
- Subjects
- Animals, Crosses, Genetic, Erythropoiesis, Feasibility Studies, Female, Gene Expression Profiling, Genes, Lethal, Genetic Vectors therapeutic use, Globins analysis, Humans, Iron Overload etiology, Iron Overload prevention & control, Liver cytology, Liver embryology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Radiation Chimera, Recombinant Fusion Proteins blood, Recombinant Fusion Proteins genetics, Transduction, Genetic, beta-Thalassemia blood, beta-Thalassemia genetics, Disease Models, Animal, Genetic Therapy, Globins genetics, Hematopoietic Stem Cell Transplantation, beta-Thalassemia therapy
- Abstract
Patients affected by beta-thalassemia major require lifelong transfusions because of insufficient or absent production of the beta chain of hemoglobin (Hb). A minority of patients are cured by allogeneic bone marrow transplantation. In the most severe of the hitherto available mouse models of beta-thalassemia, a model for human beta-thalassemia intermedia, we previously demonstrated that globin gene transfer in bone marrow cells is curative, stably raising Hb levels from 8.0-8.5 to 11.0-12.0 g/dL in long-term chimeras. To fully assess the therapeutic potential of gene therapy in the context of a lethal anemia, we now have created an adult model of beta(0)-thalassemia major. In this novel model, mice engrafted with beta-globin-null (Hbb(th3/th3)) fetal liver cells succumb to ineffective erythropoiesis within 60 days. These mice rapidly develop severe anemia (2-4 g/dL), massive splenomegaly, extramedullary hematopoiesis (EMH), and hepatic iron overload. Remarkably, most mice (11 of 13) treated by lentivirus-mediated globin gene transfer were rescued. Long-term chimeras with an average 1.0-2.4 copies of the TNS9 vector in their hematopoietic and blood cells stably produced up to 12 g/dL chimeric Hb consisting of mu alpha(2):hu beta(2) tetramers. Pathologic analyses indicated that erythroid maturation was restored, while EMH and iron overload dramatically decreased. Thus, we have established an adult animal model for the most severe of the hemoglobinopathies, Cooley anemia, which should prove useful to investigate both genetic and pharmacologic treatments. Our findings demonstrate the remarkable efficacy of lentivirus-mediated globin gene transfer in treating a fulminant blood disorder and strongly support the efficacy of gene therapy in the severe hemoglobinopathies.
- Published
- 2003
- Full Text
- View/download PDF
195. Therapeutic globin gene delivery using lentiviral vectors.
- Author
-
Rivella S and Sadelain M
- Subjects
- Humans, Genetic Vectors, Globins genetics, Lentivirus genetics
- Abstract
The severe hemoglobinopathies, including beta-thalassemia major and sickle cell anemia, are candidate diseases for a genetic treatment based on the transfer of a regulated globin gene in autologous hematopoietic stem cells. Two years ago, May et al reported that an optimized beta-globin transcription unit containing multiple proximal and distal regulatory elements harbored by a recombinant lentiviral vector could efficiently integrate into murine hematopoietic stem cells and express therapeutic levels of the human beta-globin gene. Here, we review the advantages afforded by lentivirus-mediated globin gene transfer and recent studies based on this strategy.
- Published
- 2002
196. Successful treatment of murine beta-thalassemia intermedia by transfer of the human beta-globin gene.
- Author
-
May C, Rivella S, Chadburn A, and Sadelain M
- Subjects
- Animals, Bone Marrow Cells metabolism, Bone Marrow Transplantation, Erythropoiesis drug effects, Erythropoiesis genetics, Globins administration & dosage, Globins pharmacology, Hematopoiesis, Extramedullary drug effects, Hematopoietic Stem Cell Transplantation, Hematopoietic Stem Cells, Humans, Iron Overload therapy, Mice, Recombinant Fusion Proteins biosynthesis, Recombinant Fusion Proteins blood, Recombinant Fusion Proteins pharmacology, Splenomegaly drug therapy, Splenomegaly pathology, Transduction, Genetic, beta-Thalassemia complications, Genetic Therapy methods, Globins genetics, beta-Thalassemia therapy
- Abstract
The beta-thalassemias are caused by more than 200 mutations that reduce or abolish beta-globin production. The severity of the resulting anemia can lead to lifelong transfusion dependency. A genetic treatment based on globin gene transfer would require that transgene expression be erythroid specific, elevated, and sustained over time. We report here that long-term synthesis of chimeric hemoglobin (mualpha(2):hubeta(A)(2)) could be achieved in mice with beta-thalassemia intermedia following engraftment with bone marrow cells transduced with a lentiviral vector encoding the human beta-globin gene. In the absence of any posttransduction selection, the treated chimeras exhibit durably increased hemoglobin levels without diminution over 40 weeks. Ineffective erythropoiesis and extramedullary hematopoiesis (EMH) regress, as reflected by normalization of spleen size, architecture, hematopoietic colony formation, and disappearance of liver EMH. These findings establish that a sustained increase of 3 to 4 g/dL hemoglobin is sufficient to correct ineffective erythropoiesis. Hepatic iron accumulation is markedly decreased in 1-year-old chimeras, indicating persistent protection from secondary organ damage. These results demonstrate for the first time that viral-mediated globin gene transfer in hematopoietic stem cells effectively treats a severe hemoglobin disorder.
- Published
- 2002
- Full Text
- View/download PDF
197. Therapeutic haemoglobin synthesis in beta-thalassaemic mice expressing lentivirus-encoded human beta-globin.
- Author
-
May C, Rivella S, Callegari J, Heller G, Gaensler KM, Luzzatto L, and Sadelain M
- Subjects
- Animals, Bone Marrow Transplantation, Cell Line, Female, Gene Transfer Techniques, Genetic Vectors, Globins biosynthesis, HIV-1 genetics, Humans, Male, Mice, Recombinant Proteins genetics, Recombinant Proteins pharmacology, Transduction, Genetic, beta-Thalassemia metabolism, Genetic Therapy, Globins genetics, Hemoglobins biosynthesis, Lentivirus genetics, beta-Thalassemia therapy
- Abstract
The stable introduction of a functional beta-globin gene in haematopoietic stem cells could be a powerful approach to treat beta-thalassaemia and sickle-cell disease. Genetic approaches aiming to increase normal beta-globin expression in the progeny of autologous haematopoietic stem cells might circumvent the limitations and risks of allogeneic cell transplants. However, low-level expression, position effects and transcriptional silencing hampered the effectiveness of viral transduction of the human beta-globin gene when it was linked to minimal regulatory sequences. Here we show that the use of recombinant lentiviruses enables efficient transfer and faithful integration of the human beta-globin gene together with large segments of its locus control region. In long-term recipients of unselected transduced bone marrow cells, tetramers of two murine alpha-globin and two human betaA-globin molecules account for up to 13% of total haemoglobin in mature red cells of normal mice. In beta-thalassaemic heterozygous mice higher percentages are obtained (17% to 24%), which are sufficient to ameliorate anaemia and red cell morphology. Such levels should be of therapeutic benefit in patients with severe defects in haemoglobin production.
- Published
- 2000
- Full Text
- View/download PDF
198. The cHS4 insulator increases the probability of retroviral expression at random chromosomal integration sites.
- Author
-
Rivella S, Callegari JA, May C, Tan CW, and Sadelain M
- Subjects
- 3' Untranslated Regions genetics, 3T3 Cells, 5' Untranslated Regions genetics, Animals, Cell Line, DNA Methylation, Deoxyribonuclease I metabolism, Flow Cytometry, Gene Silencing, Gene Transfer Techniques, Globins genetics, Mice, Moloney murine leukemia virus physiology, Terminal Repeat Sequences, Transgenes, Viral Proteins genetics, Virus Replication, Genetic Vectors, Moloney murine leukemia virus genetics, Regulatory Sequences, Nucleic Acid, Viral Proteins metabolism, Virus Integration
- Abstract
Retroviruses are highly susceptible to transcriptional silencing and position effects imparted by chromosomal sequences at their integration site. These phenomena hamper the use of recombinant retroviruses as stable gene delivery vectors. As insulators are able to block promoter-enhancer interactions and reduce position effects in some transgenic animals, we examined the effect of an insulator on the expression and structure of randomly integrated recombinant retroviruses. We used the cHS4 element, an insulator from the chicken beta-like globin gene cluster, which has been shown to reduce position effects in transgenic Drosophila. A large panel of mouse erythroleukemia cells that bear a single copy of integrated recombinant retroviruses was generated without using drug selection. We show that the cHS4 increases the probability that integrated proviruses will express and dramatically decreases the level of de novo methylation of the 5' long terminal repeat. These findings support a primary role of methylation in the silencing of retroviruses and suggest that cHS4 could be useful in gene therapy applications to overcome silencing of retroviral vectors.
- Published
- 2000
- Full Text
- View/download PDF
199. Selection and mapping of replication origins from a 500-kb region of the human X chromosome and their relationship to gene expression.
- Author
-
Rivella S, Palermo B, Pelizon C, Sala C, Arrigo G, and Toniolo D
- Subjects
- Blotting, Southern, Chromosome Mapping, DNA Replication genetics, Female, Humans, Lymphocytes drug effects, Male, Molecular Sequence Data, Phytohemagglutinins pharmacology, Polymerase Chain Reaction, Sequence Analysis, DNA, Transcription, Genetic, Gene Expression Regulation, Replication Origin, X Chromosome genetics
- Abstract
In higher eukaryotes the mechanism controlling initiation of DNA replication remains largely unknown. New technologies are needed to shed light on how DNA replication initiates along the genome in specific regions. To identify the human DNA sequence requirements for initiation of replication, we developed a new method that allows selection of replication origins starting from large genomic regions of human DNA. We repeatedly isolated 15 new putative replication origins (PROs) from a human DNA region of 500 kb in which 17 genes have previously been characterized. Fine-mapping of these PROs showed that DNA replication can initiate at many specific points along actively transcribed DNA in the cell lines used for our selection. In conclusion, in this paper we describe a new method to identify PROs that suggests that the availability of initiation sites is dependent on the transcriptional state of the DNA., (Copyright 1999 Academic Press.)
- Published
- 1999
- Full Text
- View/download PDF
200. Genetic treatment of severe hemoglobinopathies: the combat against transgene variegation and transgene silencing.
- Author
-
Rivella S and Sadelain M
- Subjects
- Animals, Bone Marrow, Chimera, Dependovirus genetics, Humans, Locus Control Region, Mice, Mice, Transgenic, Gene Expression Regulation, Genetic Therapy, Hemoglobinopathies therapy, Transgenes
- Abstract
Gene addition strategies are rational approaches to the treatment of sickle cell anemia and thalassemia. The goal of such genetic treatments is to introduce a functional globin transcription unit in hematopoietic stem cells and express the transgene in a manner that is erythroid-specific, elevated, relatively constant from one cell to another, and sustained over time. Gene transfer is mediated by an expanding array of viral and nonviral vectors. High-titer retroviral vectors harboring the human beta-globin gene and the core sequences of the human beta-globin locus control region yield erythroid-specific gene expression in erythroid cell lines and in short-term murine bone marrow chimeras. However, we show that expression remains subject to position effect variegation and often decreases over time in vivo. Rather than a progressive transcriptional silencing in all cells, we ascribe the waning expression to the gradual emergence in blood of erythroid progeny derived from more and more primitive precursor cells in the months after transplantation. In our model, transgene expression is therefore determined by the integration site and the differentiation stage of the transduced cell at the time of integration. Globin expression is thus different in the progeny of a transduced erythroid progenitor cell and in the erythroid progeny of a transduced hematopoietic stem cell, reflecting the effect of flanking chromatin in differentiated cells and of chromatin remodeling at the site of integration in the progeny of multipotential cells. This model predicts that insulators and matrix attachment regions could be highly valuable to gene therapy in combination with potent transcriptional activators. When efficient gene transfer in hematopoietic stem cells is achieved at last, the challenge will be to regulate gene expression in vivo and overcome transgene variegation and transgene silencing.
- Published
- 1998
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.