Review
OPA1 (dys)functions

https://doi.org/10.1016/j.semcdb.2009.12.012Get rights and content

Abstract

Mitochondrial morphology varies according to cell type and cellular context from an interconnected filamentous network to isolated dots. This morphological plasticity depends on mitochondrial dynamics, a balance between antagonistic forces of fission and fusion. DRP1 and FIS1 control mitochondrial outer membrane fission and Mitofusins its fusion. This review focuses on OPA1, one of the few known actors of inner membrane dynamics, whose mutations provoke an optic neuropathy. Since its first identification in 2000 the characterization of the functions of OPA1 has made rapid progress thus providing numerous clues to unravel the pathogenetic mechanisms of ADOA-1.

Introduction

Mitochondria form a highly dynamic reticulum which morphology varies according to cell types and contexts and depends on continuous fission and fusion of both mitochondrial outer (OM) and inner membranes (IM). In the past decade, genetic screens in yeast led to the identification of several evolutionarily conserved proteins essential for the maintenance of mitochondrial morphology. In mammals, FIS1 and DRP1 drive mitochondrial fission whereas fusion involves the OM proteins Mitofusin-1 and -2 (MFN1 and MFN2), and the IM-located OPA1 [1]. Accumulating data show the impact of mitochondrial dynamics on mitochondrial functions and on the physiology of the cell [2]. Furthermore these processes are essential for mammalian development and are affected in neurodegenerative diseases.

In this review, we will focus on OPA1 and discuss to which extent the recent progresses in elucidating the different functions of this dynamin could help understanding the pathological mechanisms of type 1 optic atrophy (ADOA-1 OMIM 165500). ADOA-1 is a neurological disease caused by mutations of OPA1, that affects retinal ganglion cells (RGC) whose axons form the optic nerve, and leads to reduced visual acuity and possibly to blindness [3].

Section snippets

OPA1 expression

The OPA1 gene encodes a mitochondrial protein localized in the inter-membrane space (IMS) and associated to mitochondrial membranes [4], [5], [6], [7]. OPA1, and its yeast orthologues Mgm1p in Saccharomyces cerevisiae and Msp1p in Schizosaccharomyces pombe, belongs to the dynamins’ family [8], [9], [10], [11]; with which it shares three conserved regions: a GTPase domain, a middle domain and a carboxy-terminal coiled-coil domain (CC-II) also called GTPase effector domain (GED) (Fig. 1). The

OPA1, mitochondrial fusion

Loss of function of OPA1 by RNAi or gene knockout, or of Mgm1p and Msp1p, causes fragmentation of the tubular mitochondrial reticulum [5], [23], [24], [25], [26]. Conversely, over-production of this protein promotes mitochondrial elongation in cells where mitochondria are punctuated [4], [27]. Surprisingly, over-expression of the dynamin in cells with tubular mitochondria causes mitochondrial fragmentation [5], [28]. However, such cells still possess a normal mitochondrial fusion activity [27],

OPA1, apoptosis

In addition to mitochondrial fragmentation, downregulation of OPA1, or expression of pathogenic mutants, increases cell sensitivity to spontaneous and induced apoptosis [25], [30], [39]. These observations lead to propose that OPA1 functions as an anti-apoptotic protein providing link between mitochondrial dynamics and apoptosis, a subject of extensive debate. Indeed, over-expression of OPA1, by inhibiting cytochrome c release, protects cells from apoptosis induced by intrinsic stimuli [46].

OPA1, mtDNA maintenance

While Mgm1p has been identified thanks to its role in mtDNA maintenance, it is only recently that OPA1 has been linked to mtDNA stability. Missense mutations in OPA1 cause accumulation of multiple deletions in skeletal muscle. The syndrome associated to these mutations (ADOA-1 plus) is complex, consisting of a combination of dominant optic atrophy, progressive external ophtalmoplegia, peripheral neuropathy, ataxia and deafness [57], [58].

As far we know at present no rearrangement but depletion

OPA1, energetic metabolism

Considering the similarity of clinical expression between ADOA-1 and Leber Hereditary Optic Neuropathy (LHON [64]) which is mainly due to mutations in subunits of respiratory complex I, the localization of OPA1 in the IM the major place of oxidative phosphorylation, and its destructuration upon inactivation of the dynamin, it can be postulated that pathogenic OPA1 mutations could lead to energetic defects. Accordingly, RNAi depleted OPA1 cells show a severe reduction of endogenous respiration,

Animal models of ADOA-1

Mitochondrial network abnormalities, bioenergetic failure, increased apoptosis have all been proposed as possible causes of ADOA-1. Recent development of in vivo models of the disease should provide new insights in understanding the physiopathological processes leading to ADOA-1.

Two Opa1 mouse models carrying an in frame deletion of 27 amino acids or a premature stop codon in the GTPase domain have been recently published [42], [67]. Both Opa1 homozygous mouse mutants are lethal at early stage

Conclusion

Numerous data have recently accumulated on the essential role of OPA1 in mitochondria. This dynamin-related protein plays role in mitochondrial fusion, cristae structuration, apoptosis and mtDNA maintenance (Fig. 2), probably reflecting its capacity to modulate IM organization and therefore activities of IM-associated proteins. However there are still gaps in our understanding of the molecular mechanisms underlying these functions. Anti-apoptotic function of OPA1 has been related to its

Acknowledgements

We are indebted to the invaluable support from the following associations: Rétina France, AFM, AMMi, ARC, LNCC and UNADV. EG, AD, TL, AO were recipients of a fellowship from ARC LNCC and FRM.

References (80)

  • T. Misaka et al.

    Primary structure of a dynamin-related mouse mitochondrial GTPase and its distribution in brain, subcellular localization, and effect on mitochondrial morphology

    J Biol Chem

    (2002)
  • H. Chen et al.

    Disruption of fusion results in mitochondrial heterogeneity and dysfunction

    J Biol Chem

    (2005)
  • S. Duvezin-Caubet et al.

    Proteolytic processing of OPA1 links mitochondrial dysfunction to alterations in mitochondrial morphology

    J Biol Chem

    (2006)
  • H. Sesaki et al.

    Cells lacking Pcp1p/Ugo2p, a rhomboid-like protease required for Mgm1p processing, lose mtDNA and mitochondrial structure in a Dnm1p-dependent manner, but remain competent for mitochondrial fusion

    Biochem Biophys Res Commun

    (2003)
  • M. Zick et al.

    Distinct roles of the two isoforms of the dynamin-like GTPase Mgm1 in mitochondrial fusion

    FEBS Lett

    (2009)
  • S. Meeusen et al.

    Mitochondrial inner-membrane fusion and crista maintenance requires the dynamin-related GTPase Mgm1

    Cell

    (2006)
  • G. Lenaers et al.

    OPA1 functions in mitochondria and dysfunctions in optic nerve

    Int J Biochem Cell Biol

    (2009)
  • C. Frezza et al.

    OPA1 controls apoptotic cristae remodeling independently from mitochondrial fusion

    Cell

    (2006)
  • L. Scorrano et al.

    A distinct pathway remodels mitochondrial cristae and mobilizes cytochrome c during apoptosis

    Dev Cell

    (2002)
  • R. Yamaguchi et al.

    Opa1-mediated cristae opening is Bax/Bak and BH3 dependent, required for apoptosis, and independent of Bak oligomerization

    Mol Cell

    (2008)
  • L. Scorrano

    Opening the doors to cytochrome c: changes in mitochondrial shape and apoptosis

    Int J Biochem Cell Biol

    (2009)
  • R. Yamaguchi et al.

    Dynamics of mitochondrial structure during apoptosis and the enigma of Opa1

    Biochim Biophys Acta

    (2009)
  • G. Perkins et al.

    New insights into mitochondrial structure during cell death

    Exp Neurol

    (2009)
  • M. Payne et al.

    Dominant optic atrophy, sensorineural hearing loss, ptosis, and ophthalmoplegia: a syndrome caused by a missense mutation in OPA1

    Am J Ophthalmol

    (2004)
  • M.V. Alavi et al.

    Subtle neurological and metabolic abnormalities in an Opa1 mouse model of autosomal dominant optic atrophy

    Exp Neurol

    (2009)
  • M. Liesa et al.

    Mitochondrial dynamics in mammalian health and disease

    Physiol Rev

    (2009)
  • S.A. Detmer et al.

    Functions and dysfunctions of mitochondrial dynamics

    Nat Rev Mol Cell Biol

    (2007)
  • N. Ishihara et al.

    Regulation of mitochondrial morphology through proteolytic cleavage of OPA1

    EMBO J

    (2006)
  • B.A. Jones et al.

    Mitochondrial DNA maintenance in yeast requires a protein containing a region related to the GTP-binding domain of dynamin

    Genes Dev

    (1992)
  • C. Delettre et al.

    Nuclear gene OPA1, encoding a mitochondrial dynamin-related protein, is mutated in dominant optic atrophy

    Nat Genet

    (2000)
  • G.J. Praefcke et al.

    The dynamin superfamily: universal membrane tubulation and fission molecules?

    Nat Rev Mol Cell Biol

    (2004)
  • C. Delettre et al.

    Mutation spectrum and splicing variants in the OPA1 gene

    Hum Genet

    (2001)
  • A. Olichon et al.

    OPA1 alternate splicing uncouples an evolutionary conserved function in mitochondrial fusion from a vertebrate restricted function in apoptosis

    Cell Death Differ

    (2007)
  • S. Duvezin-Caubet et al.

    OPA1 processing reconstituted in yeast depends on the subunit composition of the m-AAA protease in mitochondria

    Mol Biol Cell

    (2007)
  • Z. Song et al.

    OPA1 processing controls mitochondrial fusion and is regulated by mRNA splicing, membrane potential, and Yme1L

    J Cell Biol

    (2007)
  • L. Griparic et al.

    Regulation of the mitochondrial dynamin-like protein Opa1 by proteolytic cleavage

    J Cell Biol

    (2007)
  • O. Guillery et al.

    Metalloprotease-mediated OPA1 processing is modulated by the mitochondrial membrane potential

    Biol Cell

    (2008)
  • C. Alexander et al.

    OPA1, encoding a dynamin-related GTPase, is mutated in autosomal dominant optic atrophy linked to chromosome 3q28

    Nat Genet

    (2000)
  • V.R. Akepati et al.

    Characterization of OPA1 isoforms isolated from mouse tissues

    J Neurochem

    (2008)
  • E.D. Wong et al.

    The dynamin-related GTPase, Mgm1p, is an intermembrane space protein required for maintenance of fusion competent mitochondria

    J Cell Biol

    (2000)
  • Cited by (54)

    • Neurogenetic motor disorders

      2023, Handbook of Clinical Neurology
    • Dominant optic atrophy: Culprit mitochondria in the optic nerve

      2021, Progress in Retinal and Eye Research
    • Incomplete penetrance in mitochondrial optic neuropathies

      2017, Mitochondrion
      Citation Excerpt :

      DOA presents many analogies with LHON, in terms of target tissue and pattern of RGC loss, however the main clinical difference is that onset is typically before the age of 10 and the natural history is described as a relentless, frequently stable for a long time or slowly progressive optic nerve atrophy, without evidence of spontaneous recovery of vision (Carelli et al., 2004; Yu-Wai-Man et al., 2011). Genetically, over 60% of DOA families are associated with mutations in the OPA1 gene, encoding the major fusogenic protein of inner mitochondrial membrane (Landes et al., 2010; Yu-Wai-Man et al., 2010a; Lenaers et al., 2012). The pathogenic mutations affecting the OPA1 gene can be grossly distinguished in those leading to haploinsufficiency and others, missense mutations, which possibly act through a dominant negative mechanism.

    View all citing articles on Scopus
    View full text