Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Blood ; 137(14): 1945-1958, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33512417

ABSTRACT

Although BCL-xL is critical to the survival of mature erythrocytes, it is still unclear whether other antiapoptotic molecules mediate survival during earlier stages of erythropoiesis. Here, we demonstrate that erythroid-specific Mcl1 deletion results in embryonic lethality beyond embryonic day 13.5 as a result of severe anemia caused by a lack of mature red blood cells (RBCs). Mcl1-deleted embryos exhibit stunted growth, ischemic necrosis, and decreased RBCs in the blood. Furthermore, we demonstrate that MCL-1 is only required during early definitive erythropoiesis; during later stages, developing erythrocytes become MCL-1 independent and upregulate the expression of BCL-xL. Functionally, MCL-1 relies upon its ability to prevent apoptosis to promote erythroid development because codeletion of the proapoptotic effectors Bax and Bak can overcome the requirement for MCL-1 expression. Furthermore, ectopic expression of human BCL2 in erythroid progenitors can compensate for Mcl1 deletion, indicating redundancy between these 2 antiapoptotic family members. These data clearly demonstrate a requirement for MCL-1 in promoting survival of early erythroid progenitors.


Subject(s)
Erythropoiesis , Gene Deletion , Gene Expression Regulation, Developmental , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Anemia/genetics , Anemia/pathology , Animals , Apoptosis , Cells, Cultured , Embryo Loss/genetics , Embryo Loss/pathology , Erythrocytes/pathology , Erythroid Cells/pathology , Humans , Mice, Inbred C57BL
2.
Blood Adv ; 3(15): 2342-2354, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31391167

ABSTRACT

Platelet activation requires fully functional mitochondria, which provide a vital energy source and control the life span of platelets. Previous reports have shown that both general autophagy and selective mitophagy are critical for platelet function. However, the underlying mechanisms remain incompletely understood. Here, we show that Nix, a previously characterized mitophagy receptor that plays a role in red blood cell maturation, also mediates mitophagy in platelets. Genetic ablation of Nix impairs mitochondrial quality, platelet activation, and FeCl3-induced carotid arterial thrombosis without affecting the expression of platelet glycoproteins (GPs) such as GPIb, GPVI, and αIIbß3 Metabolic analysis revealed decreased mitochondrial membrane potential, enhanced mitochondrial reactive oxygen species level, diminished oxygen consumption rate, and compromised adenosine triphosphate production in Nix -/- platelets. Transplantation of wild-type (WT) bone marrow cells or transfusion of WT platelets into Nix-deficient mice rescued defects in platelet function and thrombosis, suggesting a platelet-autonomous role (acting on platelets, but not other cells) of Nix in platelet activation. Interestingly, loss of Nix increases the life span of platelets in vivo, likely through preventing autophagic degradation of the mitochondrial protein Bcl-xL. Collectively, our findings reveal a novel mechanistic link between Nix-mediated mitophagy, platelet life span, and platelet physiopathology. Our work suggests that targeting platelet mitophagy Nix might provide new antithrombotic strategies.


Subject(s)
Blood Platelets/metabolism , Membrane Proteins/metabolism , Mitophagy , Platelet Activation , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Biomarkers , Bleeding Time , Blood Platelets/ultrastructure , Carotid Artery Thrombosis/etiology , Carotid Artery Thrombosis/metabolism , Carotid Artery Thrombosis/pathology , Cell Survival/genetics , Humans , Immunophenotyping , Membrane Proteins/genetics , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/ultrastructure , Phenotype , Platelet Activation/genetics , Platelet Function Tests , Proto-Oncogene Proteins/genetics , Tumor Suppressor Proteins/genetics
3.
Nat Commun ; 7: 12353, 2016 08 10.
Article in English | MEDLINE | ID: mdl-27507172

ABSTRACT

Hereditary porphyrias are caused by mutations in genes that encode haem biosynthetic enzymes with resultant buildup of cytotoxic metabolic porphyrin intermediates. A long-standing open question is why the same causal porphyria mutations exhibit widely variable penetrance and expressivity in different individuals. Here we show that severely affected porphyria patients harbour variant alleles in the ABCB6 gene, also known as Lan, which encodes an ATP-binding cassette (ABC) transporter. Plasma membrane ABCB6 exports a variety of disease-related porphyrins. Functional studies show that most of these ABCB6 variants are expressed poorly and/or have impaired function. Accordingly, homozygous disruption of the Abcb6 gene in mice exacerbates porphyria phenotypes in the Fech(m1Pas) mouse model, as evidenced by increased porphyrin accumulation, and marked liver injury. Collectively, these studies support ABCB6 role as a genetic modifier of porphyria and suggest that porphyrin-inducing drugs may produce excessive toxicities in individuals with the rare Lan(-) blood type.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Isoantigens/genetics , Porphyrias/genetics , Porphyrins/metabolism , ATP-Binding Cassette Transporters/metabolism , Alleles , Animals , Biological Transport/genetics , Cell Membrane/metabolism , Cohort Studies , Disease Models, Animal , Female , Heme/biosynthesis , Heme/metabolism , Humans , Isoantigens/blood , Isoantigens/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Mutation , Porphyrias/metabolism , Porphyrias/urine , Porphyrins/urine , Sequence Homology, Amino Acid , Severity of Illness Index , Exome Sequencing
4.
Blood ; 126(15): 1737-8, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26450953

ABSTRACT

In this issue of Blood, Mankelow et al link phosphatidylserine (PS) exposure in sickle erythrocytes to a physiological event in reticulocyte maturation. This discovery has implications for efforts to prevent thrombosis in sickle cell disease (SCD).


Subject(s)
Anemia, Sickle Cell/blood , Anemia, Sickle Cell/pathology , Autophagy , Erythrocytes/pathology , Phosphatidylserines/metabolism , Reticulocytes/pathology , Humans
5.
Cell Cycle ; 14(21): 3441-53, 2015.
Article in English | MEDLINE | ID: mdl-26397180

ABSTRACT

Retinoblastoma-1 (RB1), and the RB1-related proteins p107 and p130, are key regulators of the cell cycle. Although RB1 is required for normal erythroid development in vitro, it is largely dispensable for erythropoiesis in vivo. The modest phenotype caused by RB1 deficiency in mice raises questions about redundancy within the RB1 family, and the role of RB1 in erythroid differentiation. Here we show that RB1 is the major pocket protein that regulates terminal erythroid differentiation. Erythroid cells lacking all pocket proteins exhibit the same cell cycle defects as those deficient for RB1 alone. RB1 has broad repressive effects on gene transcription in erythroid cells. As a group, RB1-repressed genes are generally well expressed but downregulated at the final stage of erythroid development. Repression correlates with E2F binding, implicating E2Fs in the recruitment of RB1 to repressed genes. Merging differential and time-dependent changes in expression, we define a group of approximately 800 RB1-repressed genes. Bioinformatics analysis shows that this list is enriched for terms related to the cell cycle, but also for terms related to terminal differentiation. Some of these have not been previously linked to RB1. These results expand the range of processes potentially regulated by RB1, and suggest that a principal role of RB1 in development is coordinating the events required for terminal differentiation.


Subject(s)
Cell Lineage , Erythroid Cells/metabolism , Erythropoiesis , Retinoblastoma Protein/metabolism , Animals , Cells, Cultured , Computational Biology , Databases, Genetic , E2F Transcription Factors/genetics , E2F Transcription Factors/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Genotype , Gestational Age , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Phenotype , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/metabolism , Retinoblastoma Protein/deficiency , Retinoblastoma Protein/genetics , Retinoblastoma-Like Protein p107/genetics , Retinoblastoma-Like Protein p107/metabolism , Retinoblastoma-Like Protein p130/genetics , Retinoblastoma-Like Protein p130/metabolism , Time Factors
6.
Biochim Biophys Acta ; 1853(10 Pt B): 2775-83, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25753537

ABSTRACT

Mitochondrial autophagy (mitophagy) is a core cellular activity. In this review, we consider mitophagy and related cellular processes and discuss their significance for human disease. Strong parallels exist between mitophagy and xenophagy employed in host defense. These mechanisms converge on receptors in the innate immune system in clinically relevant scenarios. Mitophagy is part of a cellular quality control mechanism, which is implicated in degenerative disease, especially neurodegenerative disease. Furthermore, mitophagy is an aspect of cellular remodeling, which is employed during development. BNIP3 and NIX are related multi-functional outer mitochondrial membrane proteins. BNIP3 regulates mitophagy during hypoxia, whereas NIX is required for mitophagy during development of the erythroid lineage. Recent advances in the field of BNIP3- and NIX-mediated mitophagy are discussed.


Subject(s)
Autophagy/physiology , Erythroid Cells/metabolism , Membrane Proteins/metabolism , Mitochondria/metabolism , Mitophagy/physiology , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Hypoxia/physiology , Erythroid Cells/cytology , Humans , Membrane Proteins/genetics , Mitochondria/genetics , Mitochondrial Membranes/metabolism , Proto-Oncogene Proteins/genetics , Tumor Suppressor Proteins/genetics
7.
Autophagy ; 10(2): 188-91, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24389674

Subject(s)
Autophagy , Career Choice , Humans
9.
Autophagy ; 8(9): 1325-32, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22906961

ABSTRACT

Elimination of defective mitochondria is essential for the health of long-lived, postmitotic cells. To gain insight into this process, we examined programmed mitochondrial clearance in reticulocytes. BNIP3L is a mitochondrial outer membrane protein that is required for clearance. It has been suggested that BNIP3L functions by causing mitochondrial depolarization, activating autophagy, or engaging the autophagy machinery. Here we showed in mice that BNIP3L activity localizes to a small region in its cytoplasmic domain, the minimal essential region (MER). The MER is a novel sequence, which comprises three contiguous hydrophobic amino acid residues, and flanking charged residues. Mutation of the central leucine residue causes complete loss of BNIP3L activity, and prevents rescue of mitochondrial clearance. Structural bioinformatics analysis predicts that the BNIP3L cytoplasmic domain lacks stable tertiary structure, but that the MER forms an α-helix upon binding to another protein. These findings support an adaptor model of BNIP3L, centered on the MER.


Subject(s)
Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism , Reticulocytes/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Computational Biology , Hydrophobic and Hydrophilic Interactions , Leucine/metabolism , Membrane Proteins/deficiency , Mice , Mitochondrial Proteins/deficiency , Molecular Sequence Data , Protein Structure, Tertiary , Structure-Activity Relationship , bcl-X Protein/metabolism
11.
Blood ; 118(5): 1192-3, 2011 Aug 04.
Article in English | MEDLINE | ID: mdl-21816839
12.
Mol Cell ; 43(4): 572-85, 2011 Aug 19.
Article in English | MEDLINE | ID: mdl-21855797

ABSTRACT

Autophagy, the primary recycling pathway of cells, plays a critical role in mitochondrial quality control under normal growth conditions and in the response to cellular stress. The Hsp90-Cdc37 chaperone complex coordinately regulates the activity of select kinases to orchestrate many facets of the stress response. Although both maintain mitochondrial integrity, the relationship between Hsp90-Cdc37 and autophagy has not been well characterized. Ulk1, one of the mammalian homologs of yeast Atg1, is a serine-threonine kinase required for mitophagy. Here we show that the interaction between Ulk1 and Hsp90-Cdc37 stabilizes and activates Ulk1, which in turn is required for the phosphorylation and release of Atg13 from Ulk1, and for the recruitment of Atg13 to damaged mitochondria. Hsp90-Cdc37, Ulk1, and Atg13 phosphorylation are all required for efficient mitochondrial clearance. These findings establish a direct pathway that integrates Ulk1- and Atg13-directed mitophagy with the stress response coordinated by Hsp90 and Cdc37.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Autophagy/physiology , Cell Cycle Proteins/physiology , Chaperonins/physiology , HSP90 Heat-Shock Proteins/physiology , Intracellular Signaling Peptides and Proteins/physiology , Mitochondria/metabolism , Protein Serine-Threonine Kinases/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autophagy-Related Protein-1 Homolog , Autophagy-Related Proteins , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Line , Chaperonins/metabolism , Erythroid Cells/cytology , Erythroid Cells/metabolism , HEK293 Cells , HSP90 Heat-Shock Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , K562 Cells , Mice , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein Stability , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/physiology
13.
Curr Opin Hematol ; 18(3): 152-7, 2011 May.
Article in English | MEDLINE | ID: mdl-21423015

ABSTRACT

PURPOSE OF REVIEW: Reticulocyte remodeling has emerged as an important model for the understanding of vesicular trafficking and selective autophagy in mammalian cells. This review covers recent advances in our understanding of these processes in reticulocytes and the role of these processes in erythroid development. RECENT FINDINGS: Enucleation is caused by the coalescence of vesicles at the nuclear-cytoplasmic junction and microfilament contraction. Mitochondrial elimination is achieved through selective autophagy, in which mitochondria are targeted to autophagosomes, and undergo subsequent degradation and exocytosis. The mechanism involves an integral mitochondrial outer membrane protein and general autophagy pathways. Plasma membrane remodeling, and the elimination of certain intracellular organelles occur through the exosomal pathway. SUMMARY: Vesicular trafficking and selective autophagy have emerged as central processes in cellular remodeling. In reticulocytes, this includes enucleation and the elimination of all membrane-bound organelles and ribosomes. Ubiquitin-like conjugation pathways, which are required for autophagy in yeast, are not essential for mitochondrial clearance in reticulocytes. Thus, in higher eukaryotes, there appears to be redundancy between these pathways and other processes, such as vesicular nucleation. Future studies will address the relationship between autophagy and vesicular trafficking, and the significance of both for cellular remodeling.


Subject(s)
Reticulocytes/cytology , Reticulocytes/pathology , Animals , Autophagy , Erythropoiesis , Humans , Reticulocytes/metabolism
14.
Antioxid Redox Signal ; 14(10): 1959-69, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21126215

ABSTRACT

B-cell leukemia/lymphoma 2 (BCL-2)/adenovirus E1B interacting protein 3 (BNIP3) and Nip-like protein X (NIX) are atypical BCL-2 homology domain 3-only proteins involved in cell death, autophagy, and programmed mitochondrial clearance. BNIP3 and NIX cause cell death by targeting mitochondria, directly through BCL-2-associated X protein- or BCL-2-antagonist/killer-dependent mechanisms, or indirectly through an effect on calcium stores in the endoplasmic reticulum. BNIP3 and NIX also induce autophagy through an effect on mitochondrial reactive oxygen species production, or by releasing Beclin 1 from inhibitory interactions with antiapoptotic BCL-2 family proteins. BNIP3 downregulates mitochondrial mass in hypoxic cells, whereas NIX is required for mitochondrial elimination during erythroid development. BNIP3 and NIX have an emerging role in human health. Cell death mediated by BNIP3 and NIX is implicated in heart disease and ischemic injury. Cancer progression is linked to loss of the prodeath function of BNIP3, but also to induction of its prosurvival activity. Finally, BNIP3 and NIX are implicated in mitochondrial quality control, which is important in aging and degenerative disease. Elucidation of the mechanisms by which BNIP3 and NIX regulate cell death, autophagy, and mitochondrial clearance may lead to treatments for these conditions.


Subject(s)
Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , bcl-2-Associated X Protein/metabolism , Animals , Autophagy/physiology , Humans , Membrane Proteins/genetics , Mitochondria/genetics , Mitochondrial Proteins/genetics , Proto-Oncogene Proteins/genetics , Tumor Suppressor Proteins/genetics , bcl-2-Associated X Protein/genetics
15.
J Biol Chem ; 285(21): 15894-905, 2010 May 21.
Article in English | MEDLINE | ID: mdl-20236933

ABSTRACT

The human beta-globin gene is expressed at high levels in erythroid cells and regulated by proximal and distal cis-acting DNA elements, including promoter, enhancer, and a locus control region (LCR). Transcription complexes are recruited not only to the globin gene promoters but also to the LCR. Previous studies have implicated the ubiquitously expressed transcription factor USF and the tissue-restricted activator NF-E2 in the recruitment of transcription complexes to the beta-globin gene locus. Here we demonstrate that although USF is required for the efficient association of RNA polymerase II (Pol II) with immobilized LCR templates, USF and NF-E2 together regulate the association of Pol II with the adult beta-globin gene promoter. Recruitment of Pol II to the LCR occurs in undifferentiated murine erythroleukemia cells, but phosphorylation of LCR-associated Pol II at serine 5 of the C-terminal domain is mediated by erythroid differentiation and requires the activity of NF-E2. Furthermore, we provide evidence showing that USF interacts with NF-E2 in erythroid cells. The data provide mechanistic insight into how ubiquitous and tissue-restricted transcription factors cooperate to regulate the recruitment and activity of transcription complexes in a tissue-specific chromatin domain.


Subject(s)
Genetic Loci/physiology , Locus Control Region/physiology , NF-E2 Transcription Factor, p45 Subunit/metabolism , RNA Polymerase II/metabolism , beta-Globins/biosynthesis , Animals , Cell Line, Tumor , Humans , Mice , NF-E2 Transcription Factor, p45 Subunit/genetics , Organ Specificity/physiology , Phosphorylation/physiology , Protein Structure, Tertiary , RNA Polymerase II/genetics , Transcription, Genetic/physiology , beta-Globins/genetics
16.
Autophagy ; 6(3): 405-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20200480

ABSTRACT

Mitochondria are the primary site of energy production in animal cells. In mitochondria, the flow of electrons through the electron transport chain creates a potential difference across the inner membrane, which is utilized for ATP production. However, due to inherent inefficiencies in electron transport, reactive oxygen species are also produced, which damage mitochondrial proteins and nucleic acids, and impair mitochondrial function. Decreased mitochondrial function causes increased reactive oxygen species generation, a decline in cellular function, and potentially cell death. Therefore, to maintain cellular homeostasis, mechanisms have evolved to selectively eliminate defective mitochondria. Mitochondria are constantly undergoing cycles of fission and fusion, and this process appears to have a role in mitochondrial quality control. Following fission, daughter mitochondria are produced, which can differ in their membrane polarization. Depolarized mitochondria are less likely to undergo subsequent fusion, and more likely to undergo autophagic clearance. As would be predicted, given the potential for cytochrome c release, depolarization is a powerful stimulus for mitochondrial clearance. Depolarization causes recruitment of the E3 ubiquitin ligase Parkin to mitochondria, which is required for their subsequent engulfment by autophagosomes. Macroautophagy pathways also appear to have a role, as hepatocytes deficient for the E1-like enzyme Atg7 accumulate abnormal mitochondria. Finally, recent studies in a developmental model have yielded insight into this process. Newly formed erythrocytes, also known as reticulocytes, eliminate their entire cohort of mitochondria during development. This process depends on the mitochondrial protein NIX, is partially dependent on autophagy, and is independent of mitochondrial depolarization. Here we describe the use of reticulocytes to study mitochondrial clearance.


Subject(s)
Autophagy/physiology , Mitochondria/metabolism , Reticulocytes/cytology , Animals , Cells, Cultured , Flow Cytometry/methods , Mice
17.
EMBO Rep ; 11(1): 45-51, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20010802

ABSTRACT

Autophagy is the cellular homeostatic pathway that delivers large cytosolic materials for degradation in the lysosome. Recent evidence indicates that autophagy mediates selective removal of protein aggregates, organelles and microbes in cells. Yet, the specificity in targeting a particular substrate to the autophagy pathway remains poorly understood. Here, we show that the mitochondrial protein Nix is a selective autophagy receptor by binding to LC3/GABARAP proteins, ubiquitin-like modifiers that are required for the growth of autophagosomal membranes. In cultured cells, Nix recruits GABARAP-L1 to damaged mitochondria through its amino-terminal LC3-interacting region. Furthermore, ablation of the Nix:LC3/GABARAP interaction retards mitochondrial clearance in maturing murine reticulocytes. Thus, Nix functions as an autophagy receptor, which mediates mitochondrial clearance after mitochondrial damage and during erythrocyte differentiation.


Subject(s)
Autophagy/physiology , Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Autophagy-Related Protein 8 Family , Binding Sites , Blotting, Western , COS Cells , Cells, Cultured , Chlorocebus aethiops , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/genetics , Molecular Sequence Data , Protein Binding , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Receptors, GABA-A/metabolism , Reticulocytes/cytology , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Substrate Specificity , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/metabolism
18.
Autophagy ; 5(7): 1064-5, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19713771

ABSTRACT

Erythrocyte formation involves the elimination of mitochondria at the reticulocyte stage of development. Nix(-/-) reticulocytes fail to eliminate their mitochondria at this step due to a defect in the targeting of mitochondria to autophagosomes. To determine the role of autophagy in this process, we generated Atg7(-/-) transplant mice. Atg7(-/-) reticulocytes exhibit a partial defect in mitochondrial clearance, demonstrating that there are both autophagy-dependent and -independent mechanisms of mitochondrial clearance. We used Atg7(-/-) autophagy-defective reticulocytes to study temporal events in mitochondrial clearance. Mitochondrial depolarization precedes elimination, but in Atg7(-/-) reticulocytes the depolarization event is markedly delayed. Since Atg7 regulates autophagosome formation, we infer that mitochondrial depolarization occurs downstream of autophagosome formation in reticulocytes. We propose that there are two mechanisms of mitochondrial clearance: one that is triggered by mitochondrial depolarization, and a second NIX-dependent mechanism, which is not. The NIX-dependent mechanism remains to be elucidated.


Subject(s)
Autophagy/physiology , Mitochondria/metabolism , Reticulocytes/physiology , Animals , Autophagy-Related Protein 7 , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Reticulocytes/cytology
19.
Blood ; 114(1): 157-64, 2009 Jul 02.
Article in English | MEDLINE | ID: mdl-19417210

ABSTRACT

Mitochondrial clearance is a well recognized but poorly understood biologic process, and reticulocytes, which undergo programmed mitochondrial clearance, provide a useful model to study this phenomenon. At the ultrastructural level, mitochondrial clearance resembles an autophagy-related process; however, the role of autophagy in mitochondrial clearance has not been established. Here we provide genetic evidence that autophagy pathways, initially identified in yeast, are involved in mitochondrial clearance from reticulocytes. Atg7 is an autophagy protein and an E1-like enzyme, which is required for the activity of dual ubiquitin-like conjugation pathways. Atg7 is required for the conjugation of Atg12 to Atg5, and Atg8 to phosphatidylethanolamine (PE), and is essential for autophagosome formation. In the absence of Atg7, mitochondrial clearance from reticulocytes is diminished but not completely blocked. Mammalian homologs of Atg8 are unmodified in Atg7(-/-) erythroid cells, indicating that canonical autophagy pathways are inactive. Thus, mitochondrial clearance is regulated by both autophagy-dependent and -independent mechanisms. In addition, mitochondria, which depolarize in wild-type cells before elimination, remain polarized in Atg7(-/-) reticulocytes in culture. This suggests that mitochondrial depolarization is a consequence rather than a cause of autophagosome formation in reticulocytes.


Subject(s)
Microtubule-Associated Proteins/physiology , Mitochondria/physiology , Reticulocytes/cytology , Reticulocytes/physiology , Animals , Autophagy , Autophagy-Related Protein 7 , Base Sequence , Cell Differentiation , DNA Primers/genetics , Erythropoiesis , Fetal Tissue Transplantation , Hepatocytes/transplantation , In Vitro Techniques , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/genetics , Mitochondria/ultrastructure , Ubiquitination
20.
Methods Enzymol ; 452: 227-45, 2009.
Article in English | MEDLINE | ID: mdl-19200886

ABSTRACT

Mitochondria are the site of oxidative phosphorylation in animal cells and a primary target of reactive oxygen species-mediated damage. To prevent the accumulation of damaged mitochondria, mammalian cells have evolved strategies for their elimination. Autophagy is one means for the controlled elimination of mitochondria; however, although there has been considerable progress in defining the requirements for nonselective autophagy, relatively little is known about the genes that regulate selective autophagy of organelles. To improve our understanding of mitochondrial autophagy in mammals, we have undertaken a genetic analysis of mitochondrial clearance in murine reticulocytes. Reticulocytes provide an ideal model to study this process, because mitochondria are rapidly cleared from reticulocytes during normal development through an autophagy-related process. Here we describe several methods for monitoring mitochondrial clearance and autophagy in reticulocytes, and show that in reticulocytes these processes require genes involved in both nonselective and selective autophagy.


Subject(s)
Autophagy/physiology , Mitochondria/metabolism , Reticulocytes/metabolism , Animals , Flow Cytometry , Immunoblotting , Mice , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Mitochondria/ultrastructure , Reticulocytes/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...