14
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Inducing mitophagy in diabetic platelets protects against severe oxidative stress

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Diabetes mellitus ( DM) is a growing international concern. Considerable mortality and morbidity associated with diabetes mellitus arise predominantly from thrombotic cardiovascular events. Oxidative stress‐mediated mitochondrial damage contributes significantly to enhanced thrombosis in DM. A basal autophagy process has recently been described as playing an important role in normal platelet activation. We now report a substantial mitophagy induction (above basal autophagy levels) in diabetic platelets, suggesting alternative roles for autophagy in platelet pathology. Using a combination of molecular, biochemical, and imaging studies on human DM platelets, we report that platelet mitophagy induction serves as a platelet protective mechanism that responds to oxidative stress through JNK activation. By removing damaged mitochondria (mitophagy), phosphorylated p53 is reduced, preventing progression to apoptosis, and preserving platelet function. The absence of mitophagy in DM platelets results in failure to protect against oxidative stress, leading to increased thrombosis. Surprisingly, this removal of damaged mitochondria does not require contributions from transcription, as platelets lack a nucleus. The considerable energy and resources expended in “prepackaging” the complex mitophagy machinery in a short‐lived normal platelet support a critical role, in anticipation of exposure to oxidative stress.

          Related collections

          Most cited references45

          • Record: found
          • Abstract: found
          • Article: not found

          Regulation of autophagy by cytoplasmic p53.

          Multiple cellular stressors, including activation of the tumour suppressor p53, can stimulate autophagy. Here we show that deletion, depletion or inhibition of p53 can induce autophagy in human, mouse and nematode cells subjected to knockout, knockdown or pharmacological inhibition of p53. Enhanced autophagy improved the survival of p53-deficient cancer cells under conditions of hypoxia and nutrient depletion, allowing them to maintain high ATP levels. Inhibition of p53 led to autophagy in enucleated cells, and cytoplasmic, not nuclear, p53 was able to repress the enhanced autophagy of p53(-/-) cells. Many different inducers of autophagy (for example, starvation, rapamycin and toxins affecting the endoplasmic reticulum) stimulated proteasome-mediated degradation of p53 through a pathway relying on the E3 ubiquitin ligase HDM2. Inhibition of p53 degradation prevented the activation of autophagy in several cell lines, in response to several distinct stimuli. These results provide evidence of a key signalling pathway that links autophagy to the cancer-associated dysregulation of p53.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Microtubule-associated protein 1 light chain 3 (LC3) interacts with Bnip3 protein to selectively remove endoplasmic reticulum and mitochondria via autophagy.

            Autophagy plays an important role in cellular quality control and is responsible for removing protein aggregates and dysfunctional organelles. Bnip3 is an atypical BH3-only protein that is known to cause mitochondrial dysfunction and cell death. Interestingly, Bnip3 can also protect against cell death by inducing mitochondrial autophagy. The mechanism for this process, however, remains poorly understood. Bnip3 contains a C-terminal transmembrane domain that is essential for homodimerization and proapoptotic function. In this study, we show that homodimerization of Bnip3 is also a requirement for induction of autophagy. Several Bnip3 mutants that do not interfere with its mitochondrial localization but disrupt homodimerization failed to induce autophagy in cells. In addition, we discovered that endogenous Bnip3 is localized to both mitochondria and the endoplasmic reticulum (ER). To investigate the effects of Bnip3 at mitochondria or the ER on autophagy, Bnip3 was targeted specifically to each organelle by substituting the Bnip3 transmembrane domain with that of Acta or cytochrome b(5). We found that Bnip3 enhanced autophagy in cells from both sites. We also discovered that Bnip3 induced removal of both ER (ERphagy) and mitochondria (mitophagy) via autophagy. The clearance of these organelles was mediated in part via binding of Bnip3 to LC3 on the autophagosome. Although ablation of the Bnip3-LC3 interaction by mutating the LC3 binding site did not impair the prodeath activity of Bnip3, it significantly reduced both mitophagy and ERphagy. Our data indicate that Bnip3 regulates the apoptotic balance as an autophagy receptor that induces removal of both mitochondria and ER.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              PINK1 deficiency impairs mitochondrial homeostasis and promotes lung fibrosis.

              Although aging is a known risk factor for idiopathic pulmonary fibrosis (IPF), the pathogenic mechanisms that underlie the effects of advancing age remain largely unexplained. Some age-related neurodegenerative diseases have an etiology that is related to mitochondrial dysfunction. Here, we found that alveolar type II cells (AECIIs) in the lungs of IPF patients exhibit marked accumulation of dysmorphic and dysfunctional mitochondria. These mitochondrial abnormalities in AECIIs of IPF lungs were associated with upregulation of ER stress markers and were recapitulated in normal mice with advancing age in response to stimulation of ER stress. We found that impaired mitochondria in IPF and aging lungs were associated with low expression of PTEN-induced putative kinase 1 (PINK1). Knockdown of PINK1 expression in lung epithelial cells resulted in mitochondria depolarization and expression of profibrotic factors. Moreover, young PINK1-deficient mice developed similarly dysmorphic, dysfunctional mitochondria in the AECIIs and were vulnerable to apoptosis and development of lung fibrosis. Our data indicate that PINK1 deficiency results in swollen, dysfunctional mitochondria and defective mitophagy, and promotes fibrosis in the aging lung.
                Bookmark

                Author and article information

                Journal
                EMBO Mol Med
                EMBO Mol Med
                10.1002/(ISSN)1757-4684
                EMMM
                embomm
                EMBO Molecular Medicine
                John Wiley and Sons Inc. (Hoboken )
                1757-4676
                1757-4684
                24 May 2016
                July 2016
                : 8
                : 7 ( doiID: 10.1002/emmm.v8.7 )
                : 779-795
                Affiliations
                [ 1 ] Section of Cardiovascular Medicine Department of Internal Medicine Yale Cardiovascular Research CenterYale University School of Medicine New Haven CTUSA
                [ 2 ] Departments of Neurology and Neurobiology, Cellular Neuroscience, Neurodegeneration and Repair Program Departments of Neurology and NeurobiologyYale University School of Medicine New Haven CTUSA
                [ 3 ] Section of Endocrinology & MetabolismYale University School of Medicine New Haven CTUSA
                [ 4 ] Department of Medicine Section of Pulmonary Critical Care and Sleep MedicineYale University School of Medicine New Haven CTUSA
                [ 5 ] Guangzhou Institute of PediatricsGuangzhou Women and Children's Medical Centre Guangzhou Medical University GuangzhouChina
                Author notes
                [*] [* ] Corresponding author. Tel: +1 203 737 5583; Fax: +1 203 737 6118; E‐mail: waiho.tang@ 123456yale.edu

                Corresponding author. Tel: +1 203 737 5583; Fax: +1 203 737 6118; E‐mail: john.hwa@ 123456yale.edu

                Author information
                http://orcid.org/0000-0001-7366-2628
                Article
                EMMM201506046
                10.15252/emmm.201506046
                4931291
                27221050
                8c2bb34b-b904-4e99-843a-f8051d6b5b29
                © 2016 The Authors. Published under the terms of the CC BY 4.0 license

                This is an open access article under the terms of the Creative Commons Attribution 4.0 License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

                History
                : 10 November 2015
                : 15 April 2016
                : 18 April 2016
                Page count
                Pages: 17
                Funding
                Funded by: NIH‐NHLBI
                Award ID: RO1‐122815
                Award ID: RO1‐HL115247
                Award ID: U54‐HL117798
                Categories
                Research Article
                Research Articles
                Custom metadata
                2.0
                emmm201506046
                July 2016
                Converter:WILEY_ML3GV2_TO_NLMPMC version:4.9.1 mode:remove_FC converted:04.07.2016

                Molecular medicine
                diabetes mellitus,mitophagy,oxidative stress,platelets,haematology,metabolism,vascular biology & angiogenesis

                Comments

                Comment on this article