Integrated protein quality-control pathways regulate free α-globin in murine β-thalassemia.

Cells remove unstable polypeptides through protein quality-control (PQC) pathways such as ubiquitin-mediated proteolysis and autophagy. In the present study, we investigated how these pathways are used in β-thalassemia, a common hemoglobinopathy in which β-globin gene mutations cause the accumulation and precipitation of cytotoxic α-globin subunits. In β-thalassemic erythrocyte precursors, free α-globin was polyubiquitinated and degraded by the proteasome. These cells exhibited enhanced proteasome activity, and transcriptional profiling revealed coordinated induction of most proteasome subunits that was mediated by the stress-response transcription factor Nrf1. In isolated thalassemic cells, short-term proteasome inhibition blocked the degradation of free α-globin. In contrast, prolonged in vivo treatment of β-thalassemic mice with the proteasome inhibitor bortezomib did not enhance the accumulation of free α-globin. Rather, systemic proteasome inhibition activated compensatory proteotoxic stress-response mechanisms, including autophagy, which cooperated with ubiquitin-mediated proteolysis to degrade free α-globin in erythroid cells. Our findings show that multiple interregulated PQC responses degrade excess α-globin. Therefore, β-thalassemia fits into the broader framework of protein-aggregation disorders that use PQC pathways as cell-protective mechanisms.

[1]  S. Keleş,et al.  Autophagy Driven by a Master Regulator of Hematopoiesis , 2011, Molecular and Cellular Biology.

[2]  J. Li,et al.  Enhancement of proteasomal function protects against cardiac proteinopathy and ischemia/reperfusion injury in mice. , 2011, The Journal of clinical investigation.

[3]  Dong-Hee Lee,et al.  Systemic Analysis of Heat Shock Response Induced by Heat Shock and a Proteasome Inhibitor MG132 , 2011, PloS one.

[4]  E. Huang,et al.  Loss of nuclear factor E2-related factor 1 in the brain leads to dysregulation of proteasome gene expression and neurodegeneration , 2011, Proceedings of the National Academy of Sciences.

[5]  B. Bain Disorders of Hemoglobin: Genetics, Pathophysiology and Clinical Management , 2011 .

[6]  James Palis,et al.  Immature erythroblasts with extensive ex vivo self-renewal capacity emerge from the early mammalian fetus. , 2011, Blood.

[7]  S. Fucharoen,et al.  Enhanced activation of autophagy in β-thalassemia/Hb E erythroblasts during erythropoiesis , 2011, Annals of Hematology.

[8]  M. Weiss,et al.  Protein quality control during erythropoiesis and hemoglobin synthesis. , 2010, Hematology/oncology clinics of North America.

[9]  S. Rivella,et al.  Hepcidin as a therapeutic tool to limit iron overload and improve anemia in β-thalassemic mice. , 2010, The Journal of clinical investigation.

[10]  E. Krüger,et al.  Proteasomal degradation is transcriptionally controlled by TCF11 via an ERAD-dependent feedback loop. , 2010, Molecular cell.

[11]  Simon Watkins,et al.  An Autophagy-Enhancing Drug Promotes Degradation of Mutant α1-Antitrypsin Z and Reduces Hepatic Fibrosis , 2010, Science.

[12]  Jun Yu,et al.  Macroautophagy modulates cellular response to proteasome inhibitors in cancer therapy. , 2010, Drug resistance updates : reviews and commentaries in antimicrobial and anticancer chemotherapy.

[13]  R. Deshaies,et al.  Transcription factor Nrf1 mediates the proteasome recovery pathway after proteasome inhibition in mammalian cells. , 2010, Molecular cell.

[14]  N. Mizushima,et al.  Methods in Mammalian Autophagy Research , 2010, Cell.

[15]  N. Chondrogianni,et al.  Nuclear Erythroid Factor 2-mediated Proteasome Activation Delays Senescence in Human Fibroblasts* , 2010, The Journal of Biological Chemistry.

[16]  D. McConkey,et al.  Proteasome inhibitors activate autophagy as a cytoprotective response in human prostate cancer cells , 2009, Oncogene.

[17]  Ji Zhang,et al.  Autophagy-dependent and -independent mechanisms of mitochondrial clearance during reticulocyte maturation , 2009, Autophagy.

[18]  D. Weatherall,et al.  Disorders of Hemoglobin: THE MOLECULAR, CELLULAR, AND GENETIC BASIS OF HEMOGLOBIN DISORDERS , 2009 .

[19]  M. Weiss,et al.  Chaperoning erythropoiesis. , 2009, Blood.

[20]  Tony Wyss-Coray,et al.  All-you-can-eat: autophagy in neurodegeneration and neuroprotection , 2009, Molecular Neurodegeneration.

[21]  H. Aburatani,et al.  Nrf1 and Nrf2 Play Distinct Roles in Activation of Antioxidant Response Element-dependent Genes* , 2008, Journal of Biological Chemistry.

[22]  H. Sandoval,et al.  Essential role for Nix in autophagic maturation of erythroid cells , 2008, Nature.

[23]  Xiao-Ming Yin,et al.  Sorting, recognition and activation of the misfolded protein degradation pathways through macroautophagy and the proteasome , 2008, Autophagy.

[24]  J. Opferman,et al.  NIX is required for programmed mitochondrial clearance during reticulocyte maturation , 2007, Proceedings of the National Academy of Sciences.

[25]  Andrew J. Gow,et al.  An erythroid chaperone that facilitates folding of α-globin subunits for hemoglobin synthesis , 2007 .

[26]  A. Gow,et al.  An erythroid chaperone that facilitates folding of alpha-globin subunits for hemoglobin synthesis. , 2007, The Journal of clinical investigation.

[27]  D. Filippov,et al.  A fluorescent broad-spectrum proteasome inhibitor for labeling proteasomes in vitro and in vivo. , 2006, Chemistry & biology.

[28]  M. Socolovsky,et al.  Suppression of Fas-FasL coexpression by erythropoietin mediates erythroblast expansion during the erythropoietic stress response in vivo. , 2006, Blood.

[29]  Terje Johansen,et al.  p62/SQSTM1 forms protein aggregates degraded by autophagy and has a protective effect on huntingtin-induced cell death , 2005, The Journal of cell biology.

[30]  Pablo Tamayo,et al.  Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[31]  S. Mandrekar,et al.  A Phase I and Pharmacologic Trial of Two Schedules of the Proteasome Inhibitor, PS-341 (Bortezomib, Velcade), in Patients with Advanced Cancer , 2005, Clinical Cancer Research.

[32]  A. Gow,et al.  Loss of α-hemoglobin–stabilizing protein impairs erythropoiesis and exacerbates β-thalassemia , 2004 .

[33]  A. Gow,et al.  Loss of alpha-hemoglobin-stabilizing protein impairs erythropoiesis and exacerbates beta-thalassemia. , 2004, The Journal of clinical investigation.

[34]  Richard Pazdur,et al.  Velcade: U.S. FDA approval for the treatment of multiple myeloma progressing on prior therapy. , 2003, The oncologist.

[35]  M. Kwak,et al.  Antioxidants Enhance Mammalian Proteasome Expression through the Keap1-Nrf2 Signaling Pathway , 2003, Molecular and Cellular Biology.

[36]  Aaron Ciechanover,et al.  The Ubiquitin Proteasome System in Neurodegenerative Diseases Sometimes the Chicken, Sometimes the Egg , 2003, Neuron.

[37]  Jiling Song,et al.  Imaging 26S proteasome activity and inhibition in living mice , 2003, Nature Medicine.

[38]  S. Koury,et al.  The effect of proteasome inhibitors on mammalian erythroid terminal differentiation. , 2002, Experimental hematology.

[39]  R. Kopito,et al.  Aggresomes, inclusion bodies and protein aggregation. , 2000, Trends in cell biology.

[40]  Wickramasinghe,et al.  Evidence that the ubiquitin proteolytic pathway is involved in the degradation of precipitated globin chains in thalassaemia , 1998, British journal of haematology.

[41]  Y. Kan,et al.  Targeted disruption of the ubiquitous CNC‐bZIP transcription factor, Nrf‐1, results in anemia and embryonic lethality in mice , 1998, The EMBO journal.

[42]  P. Detloff,et al.  A mouse model for beta 0-thalassemia. , 1995, Proceedings of the National Academy of Sciences of the United States of America.

[43]  C. Cho,et al.  A major inducer of anticarcinogenic protective enzymes from broccoli: isolation and elucidation of structure. , 1992, Proceedings of the National Academy of Sciences of the United States of America.

[44]  J. Shaeffer ATP-dependent proteolysis of hemoglobin alpha chains in beta-thalassemic hemolysates is ubiquitin-dependent. , 1988, The Journal of biological chemistry.

[45]  J. Shaeffer Turnover of excess hemoglobin alpha chains in beta-thalassemic cells is ATP-dependent. , 1983, The Journal of biological chemistry.

[46]  K. Winterhalter,et al.  Erythrocytic proteases: preferential degradation of alpha hemoglobin chains. , 1983, Acta haematologica.

[47]  D. Lester,et al.  Evidence for Increased Proteolysis in Intact β Thalassemia Erythroid Cells , 1981 .

[48]  D. Lester,et al.  Evidence for increased proteolysis in intact beta thalassemia erythroid cells. , 1981, Hemoglobin.

[49]  S. Wickramasinghe,et al.  Ultrastructural Studies of Erythropoiesis in β‐Thalassaemia Trait , 1980, British journal of haematology.

[50]  D. Loukopoulos,et al.  PROTEOLYSIS IN THALASSEMIA: STUDIES WITH PROTEASE INHIBITORS * , 1980, Annals of the New York Academy of Sciences.

[51]  S. Wickramasinghe,et al.  Ultrastructural studies of erythropoiesis in beta-thalassaemia trait. , 1980, British journal of haematology.

[52]  A. Hershko,et al.  A heat-stable polypeptide component of an ATP-dependent proteolytic system from reticulocytes. , 1978, Biochemical and biophysical research communications.

[53]  S. Wickramasinghe,et al.  Observations on the Ultrastructure of Erythropoietic Cells and Reticulum Cells in the Bone Marrow of Patients with Homozygous β‐Thalassaemia , 1975, British journal of haematology.

[54]  G. Stamatoyannopoulos,et al.  Globin synthesis in fractionated Normoblasts of beta-thalassemia heterozygotes. , 1975, The Journal of clinical investigation.

[55]  F. Gill,et al.  Free α-Globin Pool in Human Bone Marrow , 1973 .

[56]  F. Gill,et al.  Free alpha-globin pool in human bone marrow. , 1973, The Journal of clinical investigation.

[57]  J. Clegg,et al.  Haemoglobin synthesis during erythroid maturation in -thalassaemia. , 1972, Nature: New biology.

[58]  H. Lodish,et al.  Equal synthesis of - and -globin chains in erythroid precursors in heterozygous -thalassemia. , 1972, The Journal of clinical investigation.

[59]  A. Bank,et al.  Intracellular Loss of Free α Chains in β Thalassaemia , 1969, Nature.

[60]  A. Bank,et al.  Intracellular loss of free alpha chains in beta thalassemia. , 1969, Nature.

[61]  A. Bank Hemoglobin synthesis in β-thalassemia: the properties of the free α-chains , 1968 .

[62]  A. Bank Hemoglobin synthesis in beta-thalassemia: the properties of the free alpha-chains. , 1968, The Journal of clinical investigation.

[63]  R. Gunn,et al.  Thalassemia: the consequences of unbalanced hemoglobin synthesis. , 1966, The American journal of medicine.

[64]  J. Clegg,et al.  Globin Synthesis in Thalassaemia: An in vitro Study , 1965, Nature.

[65]  P. Fessas Inclusions of Hemoglobin in Erythroblasts and Erythrocytes of Thalassemia , 1963 .

[66]  P. Fessas Inclusions of hemoglobin erythroblasts and erythrocytes of thalassemia. , 1963, Blood.