19
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Defective autophagy gets to the brain

      editorial
      ,
      Oncotarget
      Impact Journals LLC
      cerebral cavernous malformations, endothelial-to-mesenchymal transition, KRIT1, LC3, MTOR, p62

      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

          Macroautophagy (herein referred to as autophagy) is an evolutionary ancient mechanism that culminates in the lysosomal degradation of useless or potentially dangerous intracellular entities, be them endogenous (like damaged organelles) or exogenous (like invading bacteria). Autophagy not only contributes to the maintenance of cellular homeostasis in physiological conditions, but also plays an essential role in adaptation to stress. In line with this notion, defective autophagy has been etiologically implicated in a wide range of human pathologies, including cancer, inflammatory disorders and neurodegenerative conditions. Recent data demonstrate that autophagic defects also contribute to the development of genetic neurovascular conditions commonly known as cerebral cavernous malformations (CCMs). CCMs (OMIM 116860, also known as cavernous angiomas or cavernomas) are neurovascular malformations that consist in clustered, aberrantly dilated and leaky capillaries lined up by a scarce endothelium and lacking normal structural components. CCMs can be inherited as a dominant autosomal disease with partial penetrance and variable expressivity or develop sporadically. Approximately 85-95% of familial CCM cases have been attributed to loss-of-function mutations in either of three genes: KRIT1, ankyrin repeat containing (KRIT1), cerebral cavernous malformation 2 (CCM2) and programmed cell death 10 (PDCD10). Mutations in unidentified genes or other hitherto unknown causes have been invoked to account for the remaining 5-15% cases of familial CCMs. In approximately 1/3 of cases, CCMs manifest clinically with moderate-to-severe symptoms including headaches, neurological deficits, seizures, strokes, and intracerebral hemorrhages. Of note, no treatment options other than the surgical resection of accessible lesions are currently available for subjects with clinically manifest CCMs [1]. Paolo Pinton and colleagues (from the University of Ferrara, Italy) have recently reported that various cell types subjected to the genetic inhibition of KRIT1 (by gene knockout or RNA interference) exhibit increased levels of two proteins that are normally processed (and degraded) by autophagy, i.e., sequestosome 1 (SQSTM1, best known as p62) and microtubule-associated protein 1 light chain 3 beta (MAP1LC3B, best known as LC3B) [2]. Such a defect was accompanied by the hyperactivation of mechanistic target of rapamycin (MTOR), a kinase with prominent autophagy-suppressing functions, and could be reversed (at least in part) by the reconstitution of KRIT1 activity as well as by the administration of two distinct pharmacological MTOR inhibitors [3, 4], namely, rapamycin and Torin 1. Similar data were obtained in Pdcd10 −/− cells. Moreover, the inability of KRIT1- deficient cells to properly process p62 and LC3 appeared to stem from defects in the late (rather than in the early) steps of autophagy, i.e., in the fusion of autophagosomes with lysosomes or in the lysosomal degradation of the autophagic cargo [5]. KRIT1-deficient cells exhibited molecular features of the so-called “endothelial-to-mesenchymal transition”, a phenotypic and biochemical shift that has previously been associated with CCMs [6]. Such molecular markers could be reversed by treating KRIT1-deficient cells with pharmacological MTOR inhibitors. Moreover, the small-interfering RNA (siRNA)-mediated depletion of an essential component of the autophagic machinery, i.e., autophagy-related 7 (ATG7), in KRIT1-proficient cells was associated with the appearance of both molecular and behavioral biomarkers of the mesenchymal state. Finally, in a mouse model of CCMs as well as in autoptic samples from CCM patients, endothelial cells from pathognomonic lesions (but not from the adjacent, normal brain) exhibited variable degrees of p62 accumulation [5]. Taken together, these data implicate defective autophagy in the pathogenesis of CCMs, and raise several important questions. First, what are the molecular mechanisms linking KRIT1 loss-of-function mutations to autophagic defects? Second, do mutations in CCM2 and PDCD10 cause similar defects or do they operate at distinct levels of the autophagic cascade? Third, is the accumulation of p62 that drives CCM pathogenesis or does this reflect a generalized impairment in endothelial cell homeostasis? Fourth, and presumably most important, would the administration of MTOR inhibitors (some of which are currently approved by regulatory agencies as immunosuppressants) [7] benefit to CCM patients? Further experiments are required to solve these incognita.

          Related collections

          Most cited references4

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

          Organelle-Specific Initiation of Autophagy.

          Autophagy constitutes a prominent mechanism through which eukaryotic cells preserve homeostasis in baseline conditions and in response to perturbations of the intracellular or extracellular microenvironment. Autophagic responses can be relatively non-selective or target a specific subcellular compartment. At least in part, this depends on the balance between the availability of autophagic substrates ("offer") and the cellular need of autophagic products or functions for adaptation ("demand"). Irrespective of cargo specificity, adaptive autophagy relies on a panel of sensors that detect potentially dangerous cues and convert them into signals that are ultimately relayed to the autophagic machinery. Here, we summarize the molecular systems through which specific subcellular compartments-including the nucleus, mitochondria, plasma membrane, reticular apparatus, and cytosol-convert homeostatic perturbations into an increased offer of autophagic substrates or an accrued cellular demand for autophagic products or functions.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Defective autophagy is a key feature of cerebral cavernous malformations

            Cerebral cavernous malformation (CCM) is a major cerebrovascular disease affecting approximately 0.3–0.5% of the population and is characterized by enlarged and leaky capillaries that predispose to seizures, focal neurological deficits, and fatal intracerebral hemorrhages. Cerebral cavernous malformation is a genetic disease that may arise sporadically or be inherited as an autosomal dominant condition with incomplete penetrance and variable expressivity. Causative loss-of-function mutations have been identified in three genes, KRIT1 (CCM1), CCM2 (MGC4607), and PDCD10 (CCM3), which occur in both sporadic and familial forms. Autophagy is a bulk degradation process that maintains intracellular homeostasis and that plays essential quality control functions within the cell. Indeed, several studies have identified the association between dysregulated autophagy and different human diseases. Here, we show that the ablation of the KRIT1 gene strongly suppresses autophagy, leading to the aberrant accumulation of the autophagy adaptor p62/SQSTM1, defective quality control systems, and increased intracellular stress. KRIT1 loss-of-function activates the mTOR-ULK1 pathway, which is a master regulator of autophagy, and treatment with mTOR inhibitors rescues some of the mole-cular and cellular phenotypes associated with CCM. Insufficient autophagy is also evident in CCM2-silenced human endothelial cells and in both cells and tissues from an endothelial-specific CCM3-knockout mouse model, as well as in human CCM lesions. Furthermore, defective autophagy is highly correlated to endothelial-to-mesenchymal transition, a crucial event that contributes to CCM progression. Taken together, our data point to a key role for defective autophagy in CCM disease pathogenesis, thus providing a novel framework for the development of new pharmacological strategies to prevent or reverse adverse clinical outcomes of CCM lesions.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Catalytic mTOR inhibitors can overcome intrinsic and acquired resistance to allosteric mTOR inhibitors

              We tested the antitumor efficacy of mTOR catalytic site inhibitor MLN0128 in models with intrinsic or acquired rapamycin-resistance. Cell lines that were intrinsically rapamycin-resistant as well as those that were intrinsically rapamycinsensitive were sensitive to MLN0128 in vitro. MLN0128 inhibited both mTORC1 and mTORC2 signaling, with more robust inhibition of downstream 4E-BP1 phosphorylation and cap-dependent translation compared to rapamycin in vitro. Rapamycin-sensitive BT474 cell line acquired rapamycin resistance (BT474 RR) with prolonged rapamycin treatment in vitro. This cell line acquired an mTOR mutation (S2035F) in the FKBP12-rapamycin binding domain; mTORC1 signaling was not inhibited by rapalogs but was inhibited by MLN0128. In BT474 RR cells, MLN0128 had significantly higher growth inhibition compared to rapamycin in vitro and in vivo. Our results demonstrate that MLN0128 may be effective in tumors with intrinsic as well as acquired rapalog resistance. mTOR mutations are a mechanism of acquired resistance in vitro; the clinical relevance of this observation needs to be further evaluated.
                Bookmark

                Author and article information

                Journal
                Oncotarget
                Oncotarget
                ImpactJ
                Oncotarget
                Impact Journals LLC
                1949-2553
                24 November 2015
                13 November 2015
                : 6
                : 37
                : 39396-39397
                Affiliations
                Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France; INSERM, U1138, Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France; Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
                Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France; INSERM, U1138, Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France; Gustave Roussy Comprehensive Cancer Institute, Villejuif, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France; Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
                Author notes
                Correspondence to: Lorenzo Galluzzi, deadoc@ 123456vodafone.it
                Correspondence to: Guido Kroemer, kroemer@ 123456orange.fr
                Article
                4741833
                26575951
                a1cb1579-9fbf-4454-8eaa-9822c97217db
                Copyright: © 2015 Galluzzi and Kroemer

                This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

                History
                : 23 October 2015
                : 10 November 2015
                Categories
                Editorial: Autophagy and Cell Death

                Oncology & Radiotherapy
                cerebral cavernous malformations,endothelial-to-mesenchymal transition,krit1,lc3,mtor,p62

                Comments

                Comment on this article