ArticlePDF AvailableLiterature Review

Clinical syndromes associated with Coenzyme Q 10 deficiency

Authors:

Abstract and Figures

Primary Coenzyme Q deficiencies represent a group of rare conditions caused by mutations in one of the genes required in its biosynthetic pathway at the enzymatic or regulatory level. The associated clinical manifestations are highly heterogeneous and mainly affect central and peripheral nervous system, kidney, skeletal muscle and heart. Genotype-phenotype correlations are difficult to establish, mainly because of the reduced number of patients and the large variety of symptoms. In addition, mutations in the same COQ gene can cause different clinical pictures. Here, we present an updated and comprehensive review of the clinical manifestations associated with each of the pathogenic variants causing primary CoQ deficiencies.
Content may be subject to copyright.
Essays in Biochemistry (2018) 62 377–398
https://doi.org/10.1042/EBC20170107
Received: 02 March 2018
Revised: 02 May 2018
Accepted: 15 May 2018
Version of Record published:
20 July 2018
Review Article
Clinical syndromes associated with Coenzyme Q10
deficiency
Mar´ıa Alc ´
azar-Fabra1, Eva Trevisson2and Gloria Brea-Calvo1
1Centro Andaluz de Biolog´
ıa del Desarrollo and CIBERER, Instituto de Salud Carlos III, Universidad Pablo de Olavide-CSIC-JA, Sevilla 41013, Spain; 2Clinical Genetics Unit,
Department of Women’s and Children’s Health, University of Padova, Padova 35128, Italy
Correspondence: Gloria Brea-Calvo (gbrecal@upo.es)
Primary Coenzyme Q deciencies represent a group of rare conditions caused by mutations
in one of the genes required in its biosynthetic pathway at the enzymatic or regulatory level.
The associated clinical manifestations are highly heterogeneous and mainly affect central
and peripheral nervous system, kidney, skeletal muscle and heart. Genotype–phenotype
correlations are difcult to establish, mainly because of the reduced number of patients and
the large variety of symptoms. In addition, mutations in the same COQ gene can cause
different clinical pictures. Here, we present an updated and comprehensive review of the
clinical manifestations associated with each of the pathogenic variants causing primary CoQ
deciencies.
Coenzyme Q structure and function
Coenzyme Q (CoQ) or ubiquinone is the only endogenously synthetized redox-active lipid that is found
in virtually all endomembranes, plasma membrane and serum lipoproteins, being especially abundant in
mitochondria. It is composed of a benzoquinone ring as a head group, and a polyisoprenoid chain, which
inserts the molecule into the phospholipid bilayer and varies in length depending on the species (Figure
1A). In humans, it has 10 isoprene units (CoQ10), 6 in Saccharomyces cerevisiae (CoQ6)andthemain
form found in mice has 9 units (CoQ9), although low amounts of CoQ10 canbealsodetectedintheir
membranes.
Soon after the first description by Cain and Morton in 1955 [1], the main function of CoQ in the mi-
tochondrial electron transport chain (mETC) was proposed by Crane et al. [2], who also demonstrated
its redox proprieties. In the mETC, CoQ is an essential mobile electron transport component, shuttling
electrons from complex I (NADH-ubiquinone oxidoreductase) or complex II (succinate-ubiquinone ox-
idoreductase) to complex III (succinate-cytochrome c oxidoreductase).
CoQ is permanently going through oxidation-reduction cycles. It can be found in a completely reduced
form (CoQH2or ubiquinol), after receiving two electrons, or in a completely oxidized form (CoQ or
ubiquinone). When, as in the mETC, this redox cycle occurs by a two-step transfer of one electron each,
a semiquinone (or semi-ubiquinone, CoQH) intermediate is produced (Figure 1B).
Computational prediction models have recently confirmed studies describing how, in the inner mi-
tochondrial membrane, CoQ is mainly either located close to the membrane–water interface, with its
relatively small head group being shadowed by the bigger polar heads of phospholipids or stabilized in
the middle of the bilayer. During the process of electron transfer, CoQ rapidly translocates from one side
to the other of the inner membrane bilayer, with a rate that varies depending on the redox state of the
molecule. This process enables the interaction with the reducing and oxidizing sites in the proteins of the
mETC complexes, located close to the membrane surfaces [3].
After the discovery of its role in the mETC, new functions have emerged for CoQ, being the electron
acceptor for different dehydrogenases. Among others, in mitochondria CoQ accepts electrons from:
(i) dihydroorotate dehydrogenase, a key enzyme for pyrimidine biosynthesis [4],
c
2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society 377
Essays in Biochemistry (2018) 62 377–398
https://doi.org/10.1042/EBC20170107
Figure 1. CoQ structure and functional integration in the mitochondrial inner membrane
(A) Chemical structure of Coenzyme Q (CoQ) and (B) redox cycle of its head group. (C) Integration of CoQ reduction by different de-
hydrogenases in the mETC; Cyt c, cytochrome c; DHODH, dihidroorotate dehydrogenase; ETF-FAD, electron transfer avoprotein;
ETF-Qase, electron transfer avoprotein cCenzyme Q reductase; G3PDH, glycerol-3-phosphate dehydrogenase; PROD, proline
dehydrogenase; SQR, sulphide-quinone oxidoreductase.
(ii) mitochondrial glycerol-3-phosphate dehydrogenase [5], a tissue-specific component of mitochon-
dria connecting glycolysis, oxidative phosphorylation and fatty acid metabolism [6],
(iii) electron transport flavoprotein dehydrogenase (ETFDH), a key enzyme involved in the fatty acid
β-oxidation and branched-chain amino acid oxidation pathways [7],
(iv) proline dehydrogenase 1, an enzyme required for proline and arginine metabolism [8],
(v) probably, from hydroxyproline dehydrogenase (or proline dehydrogenase 2), involved in the glyoxy-
late metabolism [9]
(vi) sulphide-quinone oxidoreductase [10] during sulphide detoxification, a gas modulator of relevant
cellular processes but toxic when in excess [11].
Reduced CoQ (CoQH2) generated by all these processes is efficiently reoxidized by complex III in the mETC
(Figure 1C).The ability to sustain continuous oxidation/reduction cycles makes CoQ not only a great electron carrier
for different cellular processes, but also a potent membrane antioxidant, which protects lipids, proteins and nucleic
acids from harmful oxidative damage [12,13]. In membranes, CoQH2has been shown to prevent both initiation and
propagation of lipid peroxidation [14,15] and, indirectly, to regenerate other antioxidants, such as α-tocopherol and
ascorbate [16]. The high efficiency of CoQ against oxidative stress may be related to its ubiquitous distribution, its
localization in the core of membranes and the availability of diverse dehydrogenases, able to efficiently regenerate the
molecule.
378 c
2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society
Essays in Biochemistry (2018) 62 377–398
https://doi.org/10.1042/EBC20170107
Figure 2. CoQ biosynthesis pathway
(A) Schematic model of human CoQ biosynthesis pathway. Blue arrows represent enzymatic reactions and circled numbers rep-
resent the different COQ proteins that participate in each step. Brown arrows indicate regulatory mechanisms. Circled question
mark shows currently unidentied enzymes. (B) Model of human CoQ biosynthetic complex, containing at least COQ3–COQ9
and lipids, such as CoQ itself. (Cand D) Green boxes contain 4-HB analogues, dened as unnatural CoQ precursors, which are
able to lead to CoQ production, bypassing defective COQ enzymes such as COQ6 (3,4-dihydroxybenzoate (3,4-dHB) or vanillic
acid (VA)) or COQ7 (2,4-dihydroxybenzoate (2,4-dHB). COQ9 patient broblasts can also benet from 2,4-dHB and VA; DDMQ,
demethoxy-demethyl-Coenzyme Q; DMQ, demethoxy-Coenzyme Q; DMeQ, demethyl-Coenzyme Q.
CoQ biosynthesis and regulation in eukaryotes/human
Levels of CoQ are quite stable in cells but its concentration varies among different tissues and organs, depending on
dietary conditions and age [17-20]. Although CoQ is mainly endogenously synthetized in mitochondria and then
distributed to other cell membranes [21], cells can incorporate a certain amount from dietary sources. CoQ is syn-
thesized by a set of nuclear-encoded COQ proteins, through a pathway that is not completely understood. Most of
the work on CoQ biosynthesis has been done in Saccharomyces cerevisiae, and at least 13 yeast genes (coq1coq11,
Yah 1 and Arh1) have been identified as players of this process. Information about the human pathway is very scarce,
but orthologues of almost all of these genes have been already identified [22].
4-Hydrozybenzoate (4-HB), precursor of the benzoquinone ring, is synthesized from tyrosine, phenylalanine or
also para-aminobenzoic acid (pABA) in yeast, through a poorly characterized set of reactions [23-25]. The isoprenoid
tail comes from the mevalonate pathway, which is shared with cholesterol, among other molecules, and takes place
in extra-mitochondrial membranes. This side chain is assembled by Coq1p (PDSS1 and PDSS2, acting as a heterote-
tramer, are the human orthologues), which also determines its length. Coq2p (human orthologue COQ2) condensates
head and tail and the resulting molecule undergoes subsequent modifications of the ring moiety: C5-hydroxylation
c
2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society 379
Essays in Biochemistry (2018) 62 377–398
https://doi.org/10.1042/EBC20170107
Figure 3. Organs and systems involved in CoQ primary deficiencies.
Organs and systems affected in individuals with primary CoQ deciency, associating specic clinical manifestations with the genes
involved in each one. For abbreviations refer to Table 3. For frequency of each symptom linked to a specic gene refer to Table 2.
(yeast Coq6p, human COQ6) [26], O-methylations (yeast Coq3p, human COQ3) [27,28], C1-hydroxylation and
C1-decarboxylation (unidentified), C2-methylation (yeast Coq5p, human COQ5) [29,30] and C6-hydroxylation (east
Coq7p, human COQ7) [31], but also C4-deamination (Coq6p), in the case of yeast using pABA as precursor [25].
Yah1 and Arh1 (human orthologues, FDXR and FDX2), mitochondrial ferredoxin and ferredoxin reductase, have
been shown to transfer electrons to Coq6p [32]. Mammalian pathway is still incompletely defined and significant
efforts are required in order to determine whether it coincides with the yeast one (Figure 2).
Other Coq proteins are thought to have regulatory functions. Coq8p (two human orthologues: COQ8A (or
ADCK3/CABC1) and COQ8B (or ADCK4)) displays features of an atypical kinase that possibly phosphorylates
Coq3p, Coq5p and Coq7p in yeast [33-35]. However, COQ8A/ADCK3 has recently been shown to have a more
clear ATPase activity [36] whose role in CoQ biosynthesis still needs to be further studied. Coq4p (human ortho-
logue COQ4) function has not been elucidated yet, but it seems to be required for the formation and maintenance of
the CoQ biosynthetic complex [37]. Coq9p (human orthologue COQ9) is a lipid-binding protein stabilizing Coq7p
[38,39]. Coq10p (human orthologues COQ10A and COQ10B) probably controls CoQ correct localization within the
mitochondrial membranes [40]. Coq11p is thought to be essential for CoQ synthesis in yeast, but lacks a clear human
orthologue [41]. Additionally, three other genes of the ADCK family (human ADCK1,ADCK2 and ADCK5)have
been proposed to participate in the biosynthetic process, but there is no experimental evidence for this [35,42].
It is widely accepted that yeast Coq3p–Coq9p proteins are organized in a multiprotein complex, possibly con-
taining some intermediates of the biosynthesis and CoQ itself [41,43,44]. The complex would probably optimize the
orientation of the substrates and active sites of the enzymes as well as their functional coordination [37,45-48] (Figure
380 c
2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society
Essays in Biochemistry (2018) 62 377–398
https://doi.org/10.1042/EBC20170107
2). Evidence supporting the existence of a conserved complex also in mammals has been recently reported by dif-
ferent groups through diverse approaches [24,30,36,39,49-53]. However, functional organization and regulation of
mammalian biosynthetic complex is still elusive and could be different from the yeast one.
Little is known about CoQ biosynthesis regulation, which may occur at the transcriptional, post-transcriptional and
post-translational level, or even during the assembly of the putative multisubunit complex. Transcriptionally, several
factors have emerged as possible candidates [54-56]. However, a deep study of promoters and regulation sequences of
the COQ genes is lacking currently. At the post-transcriptional level, several RNA binding proteins that modulate the
stability of COQ transcripts have also been identified [57,58]. At the post-translational level, processing by proteases,
phosphorylation and dephosphorylation have been suggested to have a role in the regulation of some COQ proteins’
activity, but only a very fragmented piece of information is currently available [34,35,59,60].
Clinical manifestations of CoQ deficiencies
CoQ deficiencies have been associated with a wide range of clinical manifestations. Patients with CoQ deficiency have
reduced levels of CoQ in tissues, which can be caused either by mutations in the genes participating in CoQ biosyn-
thesis, the so-called primary CoQ deficiencies, or by defects not directly linked CoQ biosynthesis, the secondary CoQ
deficiencies.
Primary deficiencies
Primary CoQ deficiencies are very rare conditions, usually associated with highly variable multisystemic manifes-
tations (Figure 3), and genetically caused by autosomal recessive mutations. Approximately 200 patients from 130
families have been described in the literature so far (Supplementary Table S1).
It has been estimated a worldwide total of 123,789 individuals (1 in 50,000) affected by primary CoQ deficiencies,
being only 1,665 (less than 1 in 3,000,000) due to known pathogenic variants, taking into account the frequency of
the different known or predicted pathogenic variants in given populations [118].
To date, ten genes encoding CoQ biosynthetic proteins have been shown to have pathogenic variants causing hu-
man CoQ deficiency: PDSS1,PDSS2,COQ2,COQ4,COQ5,COQ6,COQ7,COQ8A,COQ8B and COQ9 (Table
1 , Supplementary Table S1). They affect multiple organ systems in a highly variable way, including central nervous
system (CNS) (encephalopathy, seizures, cerebellar ataxia, epilepsy or intellectual disability (ID)), peripheral ner-
vous system (PNS), kidney (steroid-resistant nephrotic syndrome (SRNS)), skeletal muscle (myopathy), heart (hy-
pertrophic cardiomyopathy) and sensory system (sensorineural hearing loss (SNHL), retinopathy or optic atrophy)
(Table 2 ). While mutations in some COQ genes can affect different organs (e.g. COQ2 and COQ4), pathogenic
variants of other COQ genes show a more specific phenotype (e.g. COQ8A and COQ8B). Even more, mutations in
the same COQ gene can cause very variable clinical phenotypes with different age of onset. The age of onset may
generally range from birth to early childhood (PDSS1,PDSS2,COQ2,COQ4,COQ5,COQ6,COQ7 andCOQ9),
or from childhood to adolescence (COQ8A andCOQ8B),buttherearealsosomeadult-onsetcases(COQ2 [69];
COQ8A [99,106]; COQ8B [110]).
CNS manifestations
Central nervous system is often affected in these patients, showing a wide range of clinical manifestations, includ-
ing encephalopathy, hypotonia, seizures, dystonia, cerebellar ataxia, epilepsy, stroke-like episodes, spasticity or ID.
These symptoms may be present in patients with mutations in any of the reported COQ genes, but they are less
prominentinpatientswithpathogenicvariantsof COQ6 and COQ8B, in whom the more frequent phenotype is
renal involvement. COQ2 patients manifested early-onset nephrotic syndrome (17/22) which in some cases may
be accompanied by encephalopathy and seizures (7/22) [62, 65, 68, 71-79]. COQ4 patients generally show a severe
CNS involvement, with encephalopathy and seizures (9/14), hypotonia (10/14) and cerebellar hypoplasia (6/14); and
often a fatal outcome with death in the first days (6/14) or months (5/14) of life [80-84]. The hallmark phenotype
in COQ8A patients is slow progressive cerebellar atrophy and ataxia (43/45), associated with ID (19/45), epileptic
seizures (18/45), tremor (18/45), dysarthria (16/45), saccadic eye movements (10/45), dystonia (9/45) or spasticity
(8/45), among others [99,106,96-98, 100-105, 107-109]. The only COQ5 family described shows a phenotype similar
to COQ8A patients [85]. Some COQ8A patients (6/45) [99,97,98,102] and one COQ2 patient (1/19) [72] suffered one
stroke-like episode, which contributed significantly to deterioration of the neurological status and may explain the
heterogeneity of the functional outcome among affected siblings [97]. Some COQ2 variants have also been predicted
to increase susceptibility to adult-onset multisystem atrophy (MSA), but this issue is still under debate [69,119]. Very
few patients with mutations in PDSS1 [62,61], PDSS2 [65,63, 64, 66, 67], COQ5 [85], COQ7 [91,90] and COQ9
c
2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society 381
Essays in Biochemistry (2018) 62 377–398
https://doi.org/10.1042/EBC20170107
Tab l e 1 Pathogenic variants of COQ genes found in primary CoQ deficiency patients reported in the literature
Gene
Length
(AA) Exons RefSeq1Pathogenic variants
F (P) cDNA mutation AA modification Exon Ref.
PDSS1 415 12 exons NM 014317.4
NP 055132.2
1 (1) c.661 662insT p.Arg221Leufs* 7 [61]
1 (2) c.924T>G p.Asp308Glu 10 [62]
1 (1) c.1108A>C p.Ser370Arg 12 [61]
PDSS2 399 8 exons NM 020381.3
NP 065114.3
1 (1) c.485A>G p.His162Arg 3 [63]
1 (1) c.964C>T p.Gln322* 6 [64]
1 (1) c.1042 1148-2816 del p.? 8 [63]
2 (2) c.1145C>T p.Ser382Leu 8 [65,64]
1 (1) c.1151C>A p.Ala384Asp 8 [65]
1 (3) NK NK NK [66,67]
COQ2 2421 7 exons NM 015697.7
NP 056512.5
1 (1) c.176dupT p.Ala60Argfs*33 1 [68]
1 (1) c.382A>G p.Met128Val 1 [69,70]
3 (3) c.437G>A4p.Ser146Asn 2 [71,72,73,70]
1 (1) c.518G>A p.Arg173His 2 [65]
1 (1) c.545T>G p.Met182Arg 2 [74]
1 (1) c.590G>A p.Arg197His 3 [72,70]
7 (7) c.683A>G p.Asn228Ser 3 [72,65,75,68,70]
1 (1) c.701delT p.Leu234fs*14 4 [75,70]
1 (1) c.856C>T p.Leu286Phe 5 [65]
1 (1) c.881C>T p.Thr294Ile 5 [68]
2 (3) c.890A>G p.Tyr297Cys 5 [72, 76, 77,65,70]
1 (2) c.905C>T p.Ala302Val 5 [78,70]
1 (1) c.973A>G p.Thr325Ala 6 [68]
2 (2) c.1159C>T4p.Arg387* 7 [73,68,70]
1 (2) c.1169G>C p.Gly390Ala 7 [79]
1 (2) c.1197delT p.Asn401Ilefs*15 7 [62]
COQ4 265 7 exons NM 016035.4
NP 057119.2
1 (1) c.23 33 del TCCTCCGTCGG p.Val8Alafs*19 1 [80]
1 (2) c.155T>C p.Leu52Ser 2 [81]
1 (1) c.190C>T p.Pro64Ser 2 [81]
1 (2) c.197 198 del GCinsAA p.Arg66Gln 2 [82]
2 (3) c.202G>C p.Asp68His 2–3 [82,83]
1 (2) c.245T>A p.Leu82Gln 3 [82]
1 (1) c.311G>T p.Asp111Tyr 4 [80]
1 (1) c.356C>T p.Pro119Leu 4 [80]
1 (1) c.421C>T p.Arg141* 5 [81]
1 (1) c.433C>G p.Arg145Gly 5 [81]
1 (1) c.469C>A p.Gln157Lys 5 [83]
1 (2) c.473G>A p.Arg158Gln 5 [82]
1 (2) c.521 523 delCCA p.Thr174del 5 [81]
3 (3) c.718C>T p.Arg240Cys 7 [81,82]
1 (1) 3.9 Mb deletion of chromosome 9q34.13, including COQ4 gene [84]
COQ5 327 7 exons NM 032314.3
NP 115690.3
1 (3) 9590 pb tandem duplication of the last 4 exons of COQ5 after 1 kb of
3’-UTR (modifies the 3’-UTR) (base pair positions on Chr 12:
120,940,150–120,949,950/hg19)
[85]
COQ6 468 12 exons NM 182476.2
NP 872282.1
6 (6) c.189 191delGAA p.Lys64del 2 [86]
1 (1) c.484C>T3p.Arg162* 5 [87]
1 (1) c.564G>A3p.Trp188* 5 [87]
Continued over
382 c
2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society
Essays in Biochemistry (2018) 62 377–398
https://doi.org/10.1042/EBC20170107
Tab l e 1 Pathogenic variants of COQ genes found in primary CoQ deficiency patients reported in the literature
(Continued)
Gene
Length
(AA) Exons RefSeq1Pathogenic variants
F (P) cDNA mutation AA modification Exon Ref.
1 (1) c.686A>C p.Gln229Pro 6 [86]
2 (7) c.763G>A p.Gly255Arg 7 [87,88]
6 (6) c.782C>T p.Pro261Leu 7 [79,86]
4 (6) c.1058C>A p.Ala353Asp 9 [87,88]
1 (1) c.1078C>T p.Arg360Trp 9 [89]
1 (1) c.1154A>C p.Asp385Ala 11 [65]
2 (2) c.1235A>G3p.Tyr412Cys 11 [65,88]
1 (1) c.1341G>A p.Trp447* 11 [87,88]
1 (1) c.1383delG p.Gln461fs*478 12 [87,88]
COQ7 217 6 exons NM 016138.4
NP 057222.2
1 (1) c.332T>C (and c.308C>T) 5p.Leu111Pro (and p.
Thr103Met)
3[90]
1 (1) c.422T>A p.Val141Glu 4 [91,90]
COQ9 318 9 exons NM 020312.3
NP 064708.1
1 (1) c.521+1delG p.Ser127 Arg202del Intron 5 [92]
1 (4) c.521+2T>C p.Ser127 Arg202del Intron 5 [93]
1 (4) c.711+3G>C p.Ala203 Asp237del Intron 7 [93]
1 (1) c.730C>T p.Arg244* 7 [94,95]
COQ8A /
ADCK3
647 15 exons NM 020247.4
NP 064632.2
1 (1) c.500 521 del 22insTTG p.Gln167Leufs*36 3 [96]
1 (2) c.589-3C>G p.Leu197Valfs*20 Intron 3 [97]
1 (2) c.637C>T p.Arg213Trp 4 [97,98]
2 (3) c.811C>T p.Arg271Cys 6 [99,97]
1 (2) c.815G>T p.Gly272Val 6 [97,98]
1 (1) c.815G>A p.Gly272Asp 6 [97,98]
1 (1) c.827A>G p.Lys276Arg 6 [100]
1 (2) c.830T>C p.Leu277Pro 6 [101]
5 (7) c.895C>T p.Arg299Trp 7 [99,97,102]
1 (2) c.910G>A p.Ala304Thr 7 [99]
1 (1) c.911C>T p.Ala304Val 7 [99]
1 (1) c.993C>T6p.Lys314 Gln360del (deletion
of exon 8?)
8 [96, 97, 103]
3 (6) c.1042C>T p.Arg348* 8 [104,105]
1 (1) c.1081-1 1082 dupGTA p. Gln360 Tyr361ins* Intron 8
/Exon
9
[97]
1 (2) c.1136T>A p.Leu379* 9 [104]
1 (2) c.1228C>T p.Arg410* 10 [97]
1 (2) c.1286A>G3p.Tyr429Cys 11 [99]
1 (1) c.1358delT p.Leu453Argfs*24 11 [97]
1 (4) c.1398+2T>C7p.Asp420Trpfs*40
p.Ile467Alafs*22
11–12 [96]
1 (2) c.1506+1G>A p.Val503Metfs*21 Intron
12
[101]
1 (1) c.1511 1512delCT p.Ala504fs* 13 [106]
1 (1) c.1523T>C p.Phe508Ser 13 [97]
1 (1) c.1541A>G p.Tyr514Cys 13 [96]
1 (1) c.1645G>A p.Gly549Ser 14 [96, 97, 103]
1 (1) c.1651G>A p.Glu551Lys 14 [98]
1 (1) c.1702delG p.Glu568Argfs* 15 [100]
1 (2) c.1732T>G p.Phe578Val 15 [102]
2 (3) c.1750 1752 delACC p.Thr584del 15 [96,107]
1 (2) c.1805C>G p.Pro602Arg 15 [107]
1 (1) c.1813dupG p.Glu605Glyfs*125 15 [97,98]
1 (2) c.1844G>A p.Gly615Asp 15 [97]
1 (2) c.1844dupG p.Ser616Leufs*114 15 [108]
Continued over
c
2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society 383
Essays in Biochemistry (2018) 62 377–398
https://doi.org/10.1042/EBC20170107
Tab l e 1 Pathogenic variants of COQ genes found in primary CoQ deficiency patients reported in the literature
(Continued)
Gene
Length
(AA) Exons RefSeq1Pathogenic variants
F (P) cDNA mutation AA modification Exon Ref.
1 (1) 29 kb partial deletion of the gene including exons 3–15 (base pair
position: 227,150,977-227,195,656, hg19)
[97]
1 (1) 2.9 Mb duplication at chromosome region 1q42.11q42.13 (including
ADCK3)
[109]
COQ8B/
ADCK4
544 15 exons NM 024876.3
NP 079152.3
1 (1) c.101G>A p.Trp34* 2 [51]
4 (8) c.293T>G p.Leu98Arg 5 [110,111]
1 (2) c.449G>A p.Arg150Gln 6 [112]
3 (5) c.532C>T p.Arg178Trp 7 [51,111,113,114]
3 (4) c.645delT p.Phe215Leufs*14 8 [51,111,113]
1 (1) c.649G>A p.Ala217Thr 8 [113]
5 (5) c.737G>A p.Ser246Asn 9 [114,115]
1 (2) c.748G>A p.Asp250Asn 9 [111]
1 (1) c.748G>T p.Asp250Tyr 9 [116]
3 (4) c.748G>C8p.Asp250His 9 [117,114]
2 (3) c.759C>A p.Asn253Lys 9 [112]
1 (3) c.857A>G p.Asp286Gly 10 [51,113]
1 (1) c.929C>T p.Pro310Leu 10 [111]
1 (1) c.954 956 dupGAC p.Thr319dup 11 [51]
1 (2) c.958C>T p.Arg320Trp 11 [51,113]
1 (2) c.1027C>T p.Arg343Trp 11 [51]
5 (13) c.1199dupA p.His400Glnfs*11 13 [51,110,111,113]
7 (20) c.1339dupG p.Glu447Glyfs*10 15 [110,111,113]
1 (2) c.1356 1362 delGGGCCCT p.Gln452Hisfs* 15 [51]
2 (3) c.1430G>A p.Arg477Gln 15 [51,110,113]
1 (3) c.1447G>T p.Glu483* 15 [51,113]
1 (1) c.1468C>T p.Arg490Cys 15 [112]
1 (1) c.1493 1494 CC>AA p.Ala498Glu 15 [111]
1Reference sequences correspond to longest transcript.
2COQ2: Several initiation codons. Nomenclature is given for ATG1, but ATG4 corresponds to first amino acid (371 aa length protein) [70].
3These pathogenic variants were found in simple heterozygosis in at least one patient.
4The patient with these two mutations also carries a novel mutation in MT-ND1 (3754C>A) with 22% of heteroplasmy in peripheral blood, which
may contribute to the disease [73].
5COQ7 c.308C>T polymorphism seems to increase COQ7 protein instability and intensify the effect of the mutation. The patient also has a 1555A>G
mutation in mtDNA, not previously reported, which may contribute to the disease [90].
6Pathogenic variant c.993C>TofCOQ8A shows a conflicting protein change. Based on the sequence, it should be a synonymous change (p.Phe331
=)[96].
7COQ8A c.1398+2T>C6 variant affects a splice donor site, different splice variants are expressed: p.Asp420Trpfs*40 and p.Ile467Alafs*22 [96].
8Two siblings with this variant in homozygosis also had an homozygous mutation in NPHS1 gene (c.1339G>A; p.Glu447Lys) [117].
*stands for premature STOP codon.
Abbreviations: AA, amino acid; F(P): F, number of families with each mutation; P, number of patients with each mutation.
[92-95] have been identified to define a specific phenotype, but they presented encephalopathy (PDSS1 and COQ9),
Leigh-like syndrome (PDSS2 and COQ9), ataxia (PDSS2 and COQ5), ID (PDSS1,PDSS2,COQ5 and COQ7),
seizures (PDSS2,COQ5 and COQ9)orspasticity(PDSS2,COQ5 and COQ7).
Peripheral nervous system and sensory organs manifestations
Peripheral neuropathy has been described in two siblings with PDSS1 mutations, associated with optic atrophy
and early-onset SNHL [62]. Also, the two COQ7 patients described showed peripheral polyneuropathy, again with
SNHL and one of them with visual dysfunction [91,90]. SNHL is very frequent, especially in COQ6 patients (16/26)
[79,65,86, 87, 89], associated with SRNS in all cases, and with optic atrophy (1/18) [86]. One COQ8A patient (1/45)
384 c
2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society
Essays in Biochemistry (2018) 62 377–398
https://doi.org/10.1042/EBC20170107
Tab l e 2 Clinical manifestations in primary CoQ deficiency
Gene F P Age Clinical manifestations LA
CoQ
defi-
ciency CoQ treatment References
Onset
Last examination
(*death) CNS
PNS/sensory
organs Kidney Muscle Heart Other
Treat-
ment Effect
PDSS1 2 3 First years of
life (3)
1.5yo* (1),
14–22yo (2)
- Encephalopathy (2)2
-ID(2)
-DD(1)
- PNSN (2)
-OA(2)
- SNHL (2)
-SRNSand
ESRD (1)
-Vp(2) -LR(2)
-Obesity(2)
-HT(1)
(3) F (2)
WBC
(1)
[62,61]
PDSS2 57 <1yo (6),
2yo (1)
8mo* (2),
8yo* (1),
8–12yo (2),
NK (2)
- ARCA2 (3)
-ID(3)
-Ht(2)
-LS(1)
-Sz(1)
-Sp(1)
- Dystonia (1)
-Ny(1)
- Pyramidal
syndrome (1)
- Cerebral palsy (1)
-DD(1)
- Encephalopathy (1) 2
- SNHL (4)
-RP(2)
-Myopia(2)
-Visual
dysfunction (2)
-OA(1)
- Cataract (1)
-SRNS(7)
-ESRD(2)
- HCM (2) - Oedema (2)
-Neonatal
pneumonia (1)
-HT(1)
(2) M (1)
F(1)
Yes (2) No benefits (2) [65,63, 64, 66,
67]
COQ2 18 22 <2yo (15),
16-18yo (2),
70yo (1),
NK (1)
<2yo* (9),
2–4yo (5),
12yo (1),
23–37yo (2),
71yo (1),
NK (4)
- Encephalopathy (7) 2
-Sz(7)
-Ht(5)
-Ep(4)
-Ny(3)
-My(2)
- Dystonia (2)
- Muscle stiffness (1)
-SLL(1)
-Tr(1)
-DD(1)
-MSA(1)
-CAt (1)
-Rp(2)
-OA(1)
-RP(1)
- SRNS (17)
-ESRD(8)
-Tp(1)
-Only
proteinuria (1)
-LAM(1)
-MW(1)
- HCM (3) - Oedema (7)
-RF(4)
-LF(3)
-DM(3)
- Apnoea (2)
-RD(1)
- Cholestatic liver (1)
-HT(1)
-CLD(1)
- Hypercholestero-
laemia
(1)
(8) M (5)
F(3)
Yes (9) - No benefits (2)
- No deterioration (2)
- Renal function
restored (4)
- Neuromuscular
functions restored (1)
[69,62, 65, 68,
71-79,70]
COQ4 11 14 Birth (13),
10mo (1)
<4do* (6),
1mo–2yo* (5),
3yo (1),
18yo (1),
NK (1)
- Ht (10)
- Encephalopathy (7) 2
-CHyp(6)
-Sz(5)
-Ep(5)
-DD(4)
-ID(2)
-My(1)
-THyp(1)
- CAt (1)
-Sp(1)
-HL(1)
- PNSN (1)
- Delayed visual
maturation (1)
-Myopathy(1) -HCM(7)
- Bradycardia (4)
-HF(2)
- Cardiomegaly (1)
- HHyp (1)
-Septal
defects (1)
-RD(9)
- Apnoea (4)
-RF(2)
- Cyanosis (1)
- Dysmorphic
features (1)
- Recurrent
respirato ry
infections (1)
(9) M (6)
F(3)
Yes (3) - No benefits (1)
-Muscle
improvement (1)
- Lactic acidosis and
cardiac function
improved (1)
[80-84]
COQ5 1 3 Early
childhood (3)
14–22yo (3) - ID (3)
- ARCA2 (3)
- CAt (3)
-Dy(3)
-Ny(3)
-My(2)
-Sz(2),
-DD(2)
-Ep(1)
- SEM (1)
-Tr(1)
-Sp(1)
WBC
(3)
M(1)
Yes (3) - Improvement of
ataxia (3)
[85]
Continued over
c
2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society 385
Essays in Biochemistry (2018) 62 377–398
https://doi.org/10.1042/EBC20170107
Tab l e 2 Clinical manifestations in primary CoQ deficiency (Continued)
Gene F P Age Clinical manifestations LA
CoQ
defi-
ciency CoQ treatment References
Onset
Last examination
(*death) CNS
PNS/sensory
organs Kidney Muscle Heart Other
Treat-
ment Effect
COQ6119
263
<1.5yo (9),
2–6yo (13),
NK (4)
5–6yo* (2),
17yo* (1),
NK*(2),
0.5–11yo (14),
NK (7)
-Sz(2)
-DD(2)
- ARCA2 (1)
-ID(1)
-Pt(1)
-Exotropia(1)
-Ny(1)
- SNHL (16)
-OA(1)
- SRNS (23)
- ESRD (15)
-Only
proteinuria (1)
- MW (2) - Cardiovascular
abnormality (1)
- Dysmorphic
features (1)
Yes ( 4) - Im pr o ve d
proteinuria (3)
- Improved SNHL (1)
- Improved renal
function (1)
- Improved gro wth
retardatio n (1)
[79,65,86, 87,
89,88]
COQ7 22 <1yo (2) 6–9yo (2) - Ht (2)
-DD(2)
-ID(1)
-Sp(1)
-PNSN(2)
-SNHL(2)
-Visual
dysfunction (1)
-Kidney
dysfunction (1)
- Dysplastic
kidneys (1)
- MW (2) - HCM (1) - HT (1)
-RD(1)
-LHyp(1)
(2) M (1)
F(2)
Yes (2) - No deterioration (2) [91,90]
COQ9 3 6 Birth (6) Birth* (2),
12ho* (1),
3do* (1),
18do* (1),
2yo* (1)
-DD(5)
-Sz(2)
- Encephalopathy (2) 2
-LS(2)
- CAt (1)
- Dystonia (1)
- Opisthotonus (1)
- Muscle stiffness (1)
- Multifocal global ischaemic
events (1)
-Ht(1)
-Tp(1)
-Enlarged
kidneys (1)
- Bradycardia (2)
- Cardiomegaly (1)
- HCM (1)
- Oligohydramnios
(3)
- Apnoea (2)
-RD(1)
- Cyanosis (1)
- Reduced
haematopoiesis in
liver (1)
(4) M (1)
F(3)
Yes (3) - No benefits (2)
- Plasma lactate
reduced (1)
[92-95]
COQ8A 1
/ ADCK3
29 45 1–4yo (24),
5–11yo (13),
14–27yo(8)
22yo*(1),
26yo*(1),
3–7yo (2),
15–50yo (40),
81yo (1)
- CAt (44)
- ARCA2 (43)
- ID (19)
- Tr (18)
- Dy (16)
- My (13)
- Sz (12)
- Ep (11)
- SEM (10)
- Dystonia (9)
-Sp(8)
-Ny(6)
-SLL(6)
- Migraine (5)
-Pt(3)
-Ht(3)
- Depression (3)
-Chorea(2)
-DD(2)
- Pyramidal
syndrome (1)
- Slow ocular
pursuit (1)
- Encephalopathy (1) 2
-Strabismus(1)
-SNHL(2)
-Visual
dysfunction (1)
- Cataracts (1)
- Visual aureas
(1)
- Rod–cone
dysfunction (1)
-EI(8)
-MW(7)
-MF(3)
-LAM(3)
-Myopathy(1)
- Cardiomyopathy (1) - Recurrent
respirato ry
infections (1)
(6) M (11)
F(4)
WBC
(1)
Yes (18) - No benefits (8)
- Slight improvement
of cerebellar signs (2)
- Improvement of
tremor and
myoclonus (2)
- Improvement of
ataxia (4)
- Improvement in
motor abilities (2)
- Improvement in
fatigue
and speech (1)
- Improvement of
cognitive abilities (1)
- Stabilization of the
ataxia (1)
[99,106,96-98,
100-105,
107-109]
Continued over
386 c
2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society
Essays in Biochemistry (2018) 62 377–398
https://doi.org/10.1042/EBC20170107
Tab l e 2 Clinical manifestations in primary CoQ deficiency (Continued)
Gene F P Age Clinical manifestations LA
CoQ
defi-
ciency CoQ treatment References
Onset
Last examination
(*death) CNS
PNS/sensory
organs Kidney Muscle Heart Other
Treat-
ment Effect
COQ8B 1
/ ADCK4
38 74 <1yo–
10yo (29),
11yo–
21yo (38),
23–32yo (7)
15yo* (1),
25yo* (1),
29yo* (1),
1–9yo (6),
10–20yo (51),
21–39yo (14)
-Sz(4)
-ID(4)
- Headaches (2)
-Ep(2)
-DD(1)
- Encephalopathy (1) 2
-Autism(1)
-
Hypermetropia
- Astigmatism
(2)
-Visual
dysfunction (1)
-RP(1)
- SRNS (63)
- ESRD (45)
- CKD (17)
- Haematuria
(8)
-Only
proteinuria (8)
- Medullary
nephrocalci-
nosis (6)
-Enlarged
kidneys (1)
-MF(1)
-MW(1)
- HCM (2)
-Septal
defects (2)
-DCM(1)
- Cardiomyopathy (1)
-HF(1)
- Pericardial effusion
(1)
- Oedema (15)
- HT (10)
- Goitre (2)
-Nocturnal
enuresis (2)
- Polyuria (1)
- Polydipsia (1)
- Lupus-like
symptoms (1)
- Hypospadias (1)
- Dyspnoea (1)
- Splenomegaly (1)
- Hepatomegaly (1)
- Hypercholestero-
laemia (1)
- Dysplastic ears (1)
- Recurrent
otitis (1)
- Hypothyroidism (1)
F (5) Yes (28) - No benefits (7)
- Improvement
of proteinuria (17)
- Improvement of
oedema (1)
better physical fitness
and
- Reduced fatigue (1)
- Stabilization of
renal function (1)
-NK(2)
[51,110,111,
112-114, 116,
117]
1COQ6,COQ8B (SRNS) and COQ8A (ARCA2) variants show a very specific phenotype, but the possibility of a broader phenotype cannot be excluded, since many of the known pathogenic
variants were identified in studies that screened cohorts of patients with specific subsets of well-defined symptoms.
2Encephalopathy is defined as a wide spectrum of brain manifestations, often not described in detail.
3There are three COQ6 patients without any information.
Numbers between brackets indicate the number of patients with the specific manifestation.
*These patients died at the indicated age.
Abbreviations: CoQ deficiency (WBC, white blood cell, F, fibroblasts; M, muscle); do, days old; F, number of families; ho, hours old; LA, lactic acidosis;mo,monthsold;NK,notknown;P,
number of patients; wo, weeks old; yo, years old.
For clinical manifestations abbreviations, please refer to Table 3 and main text.
c
2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society 387
Essays in Biochemistry (2018) 62 377–398
https://doi.org/10.1042/EBC20170107
Tab l e 3 Clinical manifestations in primary CoQ deficiency
CNS PNS/sensory organs Renal Muscle Heart Other
- Autism,
- Autosomal recessive,
cerebellar ataxia 2 (ARCA2)
- Cerebellar atrophy (CAt)
- Cerebellar hypoplasia
(CHyp)
- Cerebral palsy
-Chorea
- Depression
- Developmental delay (DD)
- Dysarthria (Dy)
- Dystonia
- Encephalopathy
- Epilepsy (Ep)
- Exotropia
- Headaches
- Hypotonia (Ht)
- Intellectual deficiency (ID)
- Leigh-like syndrome (LS)
- Migraine
- Multifocal global
ischaemic events
- Muscle stiffness
- Myoclonus (My)
- Mystagmus (Ny)
- Opisthotonus
-Ptosis(Pt)
- Pyramidal syndrome
- Saccadic eye
movements (SEM)
- Seizures (Sz)
- Slow ocular pursuit
- Spasticity (Sp)
- Sporadic multisystem
atrophy (MSA),
- Strabismus
- Stroke-like lesions (SLL)
- Thalamic hypoplasia
(THyp)
-Tremor(Tr)
- Astigmatism
- Cataracts
- Delayed visual maturation
- Hearing loss (HL)
- Hypermetropia
- Myopia
- Optic nerve atrophy (OA)
- Peripheral neuropathy
(PNSN)
- Retinitis pigmentosa (RP)
- Retinopathy (Rp)
- Rod–cone dysfunction
- Sensorineural hearing
loss (SNHL)
- Visual aureas
- Visual dysfunction
- Chronic kidney disease
(CKD)
- Dysplastic kidneys
- End-stage renal disease
(ESRD)
- Enlarged kidneys
- Haematuria
- Kidney dysfunction
- Proteinuria
- Steroid resistant
nephrotic syndrome
(SRNS)
- Tubulopathy (Tp)
- Exercise intolerance (EI)
- Lipid accumulation in
muscle (LAM)
- Muscle fatigue (MF)
- Muscle weakness (MW)
- Myopathy
- Bradycardia
- Cardiomegaly
- Cardiomyopathy
- Cardiovascular
abnormality
- Dilated cardiomyopathy
(DCM)
- Heart failure(HF)
- Heart hypoplasia (HHyp)
- Hypertrophic
cardiomyopathy (HCM)
- Pericardial effusion
- Septal defects
- Valvulopathy (Vp)
- Dysmorphic features
- Oedema
- Diabetes mellitus (DM)
- Obesity
- Hypercholesterolaemia
- Goitre
- Hypothyroidism
- Respiratory distress (RD)
- Neonatal pneumonia
- Respiratory failure (RF)
- Apnoea
- Chronic lung disease (CLD)
- Recurrent respiratory
infections
-Lung hypoplasia (LHyp)
- Cyanosis
- Hypertension (HT)
- Livedo reticularis (LR)
- Dyspnoea
- Nocturnal enuresis
- Polyuria
- Polydipsia
- Liver failure (LF)
- Cholestatic liver
- Hepatomegaly
-Reduced haematopoiesis in
liver
- Lupus-like symptoms
- Splenomegaly
- Recurrent otitis
- Hypospadias
- Oligohydramnios
also showed early-onset bilateral SNHL [96, 97, 103], as well as patients with PDSS2 mutations (4/7), who mani-
fested retinitis pigmentosa (2/7) and optic atrophy (1/7), too [66,63]. One patient with COQ4 mutations (1/14) man-
ifested bilateral hearing loss as well [80]. Visual impairment was also a symptom in some patients with optic atrophy
(PDSS1 [62], PDSS2 [66,63], COQ2 [72] and COQ6 [86]), retinopathy (COQ2) [78], retinitis pigmentosa (PDSS2
[63], COQ2 [69] and COQ8B [111]) and cataracts (PDSS2 [66] and COQ8A [99]).
Renal manifestations
SRNS is frequent in primary CoQ deficiency patients, specifically in patients with pathogenic variants of COQ2,
COQ6 and COQ8B. It generally starts as proteinuria and if untreated evolves to end-stage renal disease (ESRD) within
childhood [65]. COQ2 patients displayed early-onset nephrotic syndrome (15/22) [71, 72, 76, 77,62,65,75,68], iso-
lated (9/22) or with encephalopathy and seizures (6/22), but there was also one family with onset in adolescence, slow
progression of the renal disease and mild neurological symptoms [79]. The hallmark of COQ6 pathogenic variants
is childhood-onset SNRS (23/26) associated with SNHL (16/26) [79,65,86, 87, 89,88]. COQ8B patients mainly pre-
sented with an adolescence-onset SRNS due to focal segmental glomerulosclerosis, associated with oedema (15/74)
and hypertension (10/74), which generally progressed to ESRD [51,110,111,112-114, 116, 117]. Onset of SRNS may
be before 10 years of age (29/74). Patients with PDSS1 (1/3) [61] and PDSS2 (7/7) [65,63, 64, 66, 67] mutations also
showed SRNS. One COQ9 (1/6) [95] and one COQ2 (1/22) [73] patient displayed a tubulopathy.
Muscle manifestations
Isolated myopathy has not been found in individuals with molecularly confirmed primary CoQ deficienc y. The major-
ity of the patients with a predominantly muscular phenotype have been associated with secondary CoQ deficiency.
Myopathy has been described in some patients with a multisystemic phenotype (COQ4 (1/14) [84] and COQ8A
388 c
2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society
Essays in Biochemistry (2018) 62 377–398
https://doi.org/10.1042/EBC20170107
(1/45) [100]). Other muscular manifestations include exercise intolerance (COQ8A (8/45) [96,97, 98, 104]), muscle
weakness (COQ2 (1/22) [72], COQ6 (2/26) [86], COQ7 (2/2) [91,90], COQ8A (7/45) [99,98,102,109] and COQ8B
(1/74) [117]) and muscle fatigue (COQ8A (2/45) [99,108] and COQ8B (1/74) [111]). Some muscle biopsies have
shown lipid accumulation in muscle (COQ4 (1/14) [84], COQ8A (3/45) [99,98] and COQ2 (1/22) [74]).
Cardiac manifestations
The most frequent heart manifestation is hypertrophic cardiomyopathy, often present in COQ4 patients with a pre-
natal onset (7/14) [80-82], whereas COQ2 (3/22) [71,74,73], COQ8B (2/74) [110,113,117], COQ7 (1/2) (101) and
COQ9 patients (1/6) [95] show a neonatal onset. Other less frequently reported cardiac manifestations are valvu-
lopathies (PDSS1 (2/3) [62]), heart hypoplasia (COQ4 (1/14) [81]), septal defects (COQ4 (1/14) [84] and COQ8B
(2/74) [111,112]), heart failure (COQ4 (2/14) [81,82] and COQ8B (1/74) [111]), bradycardia (COQ4 (4/14) [80-82],
COQ9 (2/6) [92,93] or pericardial effusion (COQ8B (1/74) [110,113]). However, it is questionable whether some
manifestations such as heart failure, bradycardia or pericardial effusion are primary events or are secondary mani-
festations of some other general phenomena.
Other manifestations
Less frequent clinical findings include dysmorphic features [84,87], metabolic pathologies (diabetes mellitus [62,73],
obesity [62] and hypercholesterolaemia [79,117], although the latest is often observed during SRNS, independently
of its aetiology), thyroid disease (goitre [51,113], hypothyroidism [110]), lung involvement (respiratory distress, very
frequent in COQ4 patients (9/14) [73,81,82,91,92], apnoea [78,80-82,92] or respiratory failure [72,78,73,80,81]), cir-
culatory problems (cyanosis [81,92], hypertension, livedo reticularis [62]), liver abnormalities (hepatic insufficiency
[62,74], cholestatic liver [73]), among others.
Biochemical findings
Primary CoQ deficiency patients, particularly those with neonatal onset, can show higher levels of lactate in plasma
or serum. CoQ levels in skeletal muscle biopsies or fibroblasts may be reduced [120], as well as the enzymatic activities
of complex I+III and/or II+III [121].
Pathogenesis
The pathogenesis of CoQ deficiency is complex and not completely understood. The bioenergetic defect and the
increased reactive oxygen species (ROS) production may have a crucial role. However the wide spectrum of CoQ
functions, the unclear roles of some COQ gene products and the considerable phenotypic variability, suggests that
other mechanisms contribute to the pathogenesis of the disease. In cultured cells it has been found that, while severe
CoQ deficiencies lead to great defects in energy production with no major increase in oxidative stress, mild CoQ
defects cause a significant increase in ROS production without affecting ATP production, but yielding increased cell
death levels [115]. In addition, as expected, CoQ deficiency impairs de novo pyrimidine synthesis, further contribut-
ing to disease pathogenesis [122]. CoQ deficiency cells also show increased mitophagy, being proposed as a protective
mechanism in disease pathogenesis [123], although other authors defined it as detrimental [124]. Recently, sulphide
oxidation pathway impairment has been proposed as an additional pathomechanism in primary CoQ deficiency, as
different in vivo and in vitro models of the disease show a tissue-specific defect in the metabolism of H2S, leading
to the accumulation of this molecule, which may alter protein S-sulfhydrilation, inducing changes such as vasore-
laxation, inflammation and ROS production [125]. Finally, CoQ deficiency has also been linked to development of
insulin resistance in human and mouse adipocytes, as a result of increased ROS production via complex II [126].
Genotype–phenotype correlation
Due to the small number of patients harbouring mutations in COQ genes and the wide range of clinical manifesta-
tions, it is arduous to define genotype–phenotype correlations. In fact, only a few families with pathogenic variants
of PDSS1,PDSS2,COQ5 or COQ9 have been published, being unachievable to establish any correlation. In the case
of COQ9, studies in two mouse models suggest that a key factor appears to be the different degree of impairment
of formation of the CoQ complex [50]. Even though only two patients with COQ7 mutations have been described,
there seems to be a correlation between the residual levels of CoQ (and levels of COQ7 protein) and the severity of
the disease: fibroblasts from the patient with the most severe phenotype show a drastic CoQ deficiency [91], while
the patient with the milder phenotype has a 30% decrease in CoQ levels in skin fibroblasts [90]. Interestingly, only
fibroblasts with a severe deficiency benefit from 2,4-dihydroxybenzoic acid (2,4-dHB) supplementation, while CoQ
biosynthesis was inhibited in those with the milder defect treated with 2,4-dHB.
c
2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society 389
Essays in Biochemistry (2018) 62 377–398
https://doi.org/10.1042/EBC20170107
COQ8A and COQ8B have the highest number of families with pathogenic variants reported (29 and 38), and in
neither case there is any correlation between the mutations and the clinical phenotype [97,113]. In the case of COQ2
patients (18 families described), who show the widest clinical spectrum, it has been proposed that the severity of the
disease correlates with the enzymatic residual activity and hence CoQ levels, as shown by expressing mutant proteins
inyeast[70].ItisworthtomentionthatmostoftheCOQ6 patients were diagnosed during screening for SNRS, so
there may be a reference bias in these cases [65,87,86]. To date, no other clear correlations have been observed for
COQ4 patients.
Diagnosis
ThediagnosisofprimaryCoQdeficiencyisestablishedwiththeidentificationofbiallelicpathogenicvariantsinanyof
the genes coding for one of the proteins directly involved in CoQ biosynthesis. Genome or specific gene sequencing is
performed when decreased levels of CoQ or reduced combined activities of complex I+III and II+III in mitochondria
of skeletal muscle biopsies are detected in patients [127,128]. It is important to note that biochemical analysis is not
able to distinguish between primary and secondary CoQ deficiencies [128]. Genetic identification of new pathogenic
variants is usually followed by functional validation.
CoQ levels can also be measured on plasma samples, white blood cells or skin fibroblasts obtained after skin biopsy
from patients [129]. However, there are concerns about CoQ plasma measurements for diagnosis, since it seems to
be influenced by the amount of plasma lipoproteins (carriers of CoQ in circulation) and the dietary intake. Muscle
or fibroblasts represent the preferred diagnosis tissues, although sometimes fibroblasts do not show reduction while
muscle does [130]. It has been shown that white blood cells CoQ levels alone are not reliable to diagnose primary
CoQ deficiency in the setting of nephrotic syndrome [68]. De novo synthesis can also be measured by radioactive
precursor incorporation in fibroblasts [131] which is especially useful to discriminate between primary and secondary
deficiencies. Recently, urine CoQ measurement as non-invasive approach has been proposed [132].
Management
Considering the wide clinical spectrum of this condition, any individual with a diagnosis of primary CoQ deficiency
should be assessed, in order to establish the severity of the disease. Importantly, a genetic consultation is recom-
mended for other family members and for recurrence risk of patient’s parents. Based on the genetic defect identified
in the patient, a specific follow-up should be programmed.
Being CNS manifestations very frequent, every patient with a diagnosis of CoQ deficiency should undergo peri-
odical neurological examinations, even if normal at diagnosis. In fact, the age of onset of these symptoms is highly
variable, ranging from the first hours or days of life (as in patients with COQ4 mutations),uptotheseventhdecade
of life (as in COQ2 patients with the adult-onset multisystem atrophy-like phenotype). Evaluation should include an
EEG analysis and a brain MRI. In addition, peripheral nervous system should be assessed for the possible presence
of peripheral neuropathy in patients with PDSS1 and COQ7 mutations.
Patients with mutations in PDSS1,PDSS2,COQ2,COQ6,COQ7,COQ8A and COQ8B may have eye involve-
ment due to optic atrophy, retinopathy, retinitis pigmentosa and even cataracts and should therefore be screened
at diagnosis and during the follow-up. Audiometry is necessary in COQ6 patients who almost invariably manifest
SRNS, but should be performed also in patients with mutations in PDSS1,COQ8A and COQ4 who may sometimes
manifest this phenotype.
Individuals harbouring mutations in COQ2,PDSS1,PDSS2,COQ6 and COQ8B may manifest renal involvement
with SRNS, whose onset may vary from early childhood to adolescence. Tubulopathy has been reported rarely. These
patients thus need to undergo periodical renal function tests with urine analysis for proteinuria and nephrological
evaluations for the risk of evolving to ESRD.
A cardiologist examination with echocardiogram should be performed in patients with COQ4 mutations (who may
present with a severe prenatal-onset cardiomyopathy) and should also be considered in individuals with mutations
in PDSS1 and COQ8B to exclude the presence of a valvulopathy or septal defects.
Treatment
Barriers for tissues CoQ delivery have been found due to its high molecular weight and poor aqueous solubility, but
at high doses, dietary supplementation increases CoQ levels in all tissues, including heart and brain, especially with
certain formulations [133,134]. It also increases in circulating low-density lipoproteins (LDL), where it functions as
an efficient antioxidant together with α-tocopherol [135,136]. CoQ supplementation at high doses has been demon-
strated to be effective for treatment of both primary and secondary CoQ deficiencies [137]. It is crucial to start the
390 c
2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society
Essays in Biochemistry (2018) 62 377–398
https://doi.org/10.1042/EBC20170107
supplementation as soon as possible to get favourable outcomes and to limit irreversible damage in critical tissues
such as the kidney or the CNS [127]. Different doses of CoQ have been employed for the treatment of primary CoQ
deficiencies, ranging from 5 mg/kg/day [66] to 30–50 mg/kg/day for both adults and children [138] but, in mouse
models of this condition, even higher doses (up to 200 mg/kg/day) have been used [139]. Except for COQ8A patients,
most individuals with primary forms show a good response to CoQ treatment, which is usually evident after 10–20
days [138]. Different formulations of CoQ are now available, both in the oxidized and the reduced forms, although
most of the data available have been obtained in patients treated with ubiquinone.
Alternatively to CoQ10 supplementation, some 4-HB analogues have been proposed as potential bypass molecules
with higher bioavailability than CoQ. These water-soluble CoQ head precursors would bypass enzymatic steps dis-
rupted by mutations in COQ genes, but their efficacy may differ depending on the stability of the CoQ biosynthetic
complex. Some examples are vanillic acid (VA) and 3,4-dihydroxybenzoate (3,4-dHB), able to bypass COQ6 muta-
tions, or 2,4-dHB for COQ7 defects (Figure 2C,D). The effectivity of VA and 3,4-dHB in restoring CoQ biosynthesis
has been demonstrated in coq6 yeast mutant strains expressing pathogenic versions of human COQ6 [88]. Notably,
VA also stimulates CoQ synthesis and improves cell viability in COQ9 patient fibroblasts [140]. 2,4-dHB was able to
increase CoQ levels and lifespan in Coq7 [141] and Coq9 defective mice [50], as well as to bypass the reaction in hu-
man fibroblasts with COQ7 [91,90,140] and COQ9 mutations [140]. Remarkably, the effectivity of 2,4-dHB depends
onthenatureoftheCOQ7 mutation and the residual activity of the protein [90]. It has also been reported that treat-
ment with high doses of 4-HB, thus increasing COQ2 substrate availability, restores CoQ synthesis in COQ2-deficient
cell lines, which also suggests that these enzyme variants retain some residual activity [142].
Early onset CoQ deficiencies can cause mortality in few days. We have observed that CoQ is efficiently incorporated
in different tissues by breastfeeding and placenta in mice (unpublished data). We propose treatment of pregnant
mothers of high-risk newborns (high probability of CoQ deficiency after genetic screening or due to family history)
with CoQ supplementation, in order to reduce tissue damage during embryonic/foetal development and to increase
the survival of newborns until they can be fed with supplements.
Secondary deficiencies
CoQ levels can also be reduced secondary to conditions not directly linked to CoQ biosynthesis, but related to ox-
idative phosphorylation (OXPHOS), other non-OXPHOS mitochondrial processes, or even to non-mitochondrial
functions [143]. Remarkably, secondary CoQ deficiencies are proved to be more common than primary deficiencies
[143,144], probably because of the diversity of biological functions and metabolic pathways in which CoQ is involved
in mitochondrial and non-mitochondrial membranes, highlighting the importance of CoQ homoeostasis in human
health. However, there is a lack of consistency of CoQ deficiency presence among different patients, which could sug-
gest different susceptibility to the development of secondary deficiencies among different individuals. Currently, there
is not any general explanation for this, although genetic factors, such as certain polymorphisms, have been proposed
to be involved [113,143-145]. A comprehensive analysis of muscle and fibroblasts samples from patients affected by a
wide range of mitochondrial diseases showed that secondary deficiencies were more frequent in depletion syndromes
than in any other mitochondrial disease [143], supporting previous observations [146]. The same study analysed CoQ
levels in samples of patients affected by different OXPHOS diseases, but were unable to find any difference between
them. Further studies on wider cohorts are needed in order to understand whether certain diseases are more prone to
develop secondary deficiencies than others, as well as the underlying molecular mechanism. Nonetheless, it is clear
that mitochondrial myopathies are frequently associated with CoQ secondary deficiencies [145]. Besides its reduction
in many mitochondrial OXPHOS disorders, other diseases may display secondary CoQ deficiency, including ataxia
and oculomotor apraxia syndrome (MIM #208920), multiple acyl-CoA dehydrogenase deficiency (MIM #231680),
cardiofaciocutaneous syndrome (MIM #115150), methylmalonic aciduria (# 251000), GLUT-1 deficiency syndrome
(MIM #606777), mucopolysaccharidosys type III (MIM #605270) or multisystem atrophy [143,144,147]. The mecha-
nisms underlying CoQ secondary defects remain largely unknown, but several explanations have been proposed that
are related to: (i) an increased rate of CoQ degradation due to oxidative damage caused by a non-functional respi-
ratory chain; (ii) a decrease in CoQ through the interference with the signalling pathways involved in the process of
biosynthesis; (iii) the reduction in the stability of the CoQ biosynthetic complex or (vi) a general deterioration of mi-
tochondrial function [143,144]. In addition, CoQ seems to be reduced in the process of ageing [148] and a secondary
deficiency of CoQ may be a side effect of hypercholesterolaemia treatment with statins, since both cholesterol and
CoQ share part of their biosynthetic pathways [149,150].
c
2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society 391
Essays in Biochemistry (2018) 62 377–398
https://doi.org/10.1042/EBC20170107
Of course, particular symptoms of secondary CoQ deficiencies are highly dependent on the original pathology.
Myopathies presented as muscular weakness, hypotonia, exercise intolerance or myoglobinuria are commonly re-
ported as muscular manifestations in diseases associated with secondary CoQ deficiencies. Neurological decline and
ataxia are also often reported [144,151]. It is possible that the primary disease symptoms are potentiated by the lack of
CoQ [144]. In fact, many of these patients partially improve their condition by CoQ supplementation, which supports
the importance of an early diagnosis also in these cases [151]. From the point of view of the molecular diagnosis, it is
necessary to perform a genetic analysis to distinguish between primary and secondary deficiencies [127].
Concluding remarks
The deficiency in CoQ is a genetically and clinically heterogeneous syndrome. Primary deficiency diagnosis is a great
challenge due to the number of genes involved, the poor knowledge of CoQ biosynthesis pathway and its regula-
tioninhumans,thesmallnumberofpatientsdescribedandthegreatvarietyofassociatedsymptoms.Moreover,
secondary deficiencies can be consequences of many other mitochondrial dysfunctions adding a layer of complex-
ity to the diagnosis. Observation of the clinical manifestations here described and/or the molecular identification of
potentially pathological variants of COQ genes should be complemented by the biochemical determination of CoQ
levels, biosynthesis rate if possible, and the combined enzymatic activities of complexes I+III and II+III in muscle or
fibroblast. It is important to identify potential cases as early as possible because high-dose CoQ oral supplementation
is a very effective treatment in most cases, blocking the progression of the disease.
Summary
CoQ is an endogenously synthesized lipid that is essential for the electron transport in the mito-
chondrial respiratory chain.
Primary CoQ deciencies are rare diseases caused by mutations in genes of the CoQ biosynthesis
pathway.
CoQ deciencies are characterized by reduced levels of CoQ affecting energy production and
other processes.
Primary CoQ deciencies show highly heterogeneous manifestations mainly affecting CNS, PNS,
sensory organs, kidney, skeletal muscle and heart.
Currently, it is hard to establish any genotype–phenotype correlations for these diseases, partially
due to the low amount of studied patients.
It is essential to biochemically determine CoQ deciency since supplementation has positive ther-
apeutic effects.
Acknowledgements
Authors wish to thank the patients and their families for facilitating the research here reviewed. We thank Prof. Pl´
acido Navas for
the critical revision of the manuscript.
Competing Interests
The authors declare that there are no competing interests associated with the manuscript.
Funding
This work has been partially funded by the Spanish Ministry of Health, Instituto de Salud Carlos III (ISCIII), FIS PI14-01962 and FIS
PI17-01286. M.A.-F. is a predoctoral research fellow from the Spanish Ministry of Education, Culture and Sports [FPU14/04873].
E.T. is supported by grant number [CPDA140508/14] from University of Padova.
392 c
2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society
Essays in Biochemistry (2018) 62 377–398
https://doi.org/10.1042/EBC20170107
Author Contribution
M.A.-F. exhaustively compiled the mutations and symptoms data from literature and elaborated the tables. M.A.-F. and G.B.-C.
made the gures. M.A.-F., E.T. and G.B.-C. wrote and edited the text and G.B.-C. coordinated the work.
Abbreviations
2,4-dHB, 2,4-dihydroxybenzoic acid; 3,4-dHB, 3,4-dihydroxybenzoate; 4-HB, 4-hydrozybenzoate; CNS, central nervous sys-
tem; CoQ, coenzyme Q; EEG, electroencephalography; ESRD, end-stage renal disease; ETFDH, electron transport avo-
protein dehydrogenase; HHB, hexaprenyl-hydroxybenzoate; ID, intellectual disability; LDL, low-density lipoprotein; mETC,
mitochondrial electron transport chain; MRI, magnetic resonance imaging; OXPHOS, oxidative phosphorylation; pABA,
para-aminobenzoic acid; PNS, peripheral nervous system; ROS, reactive oxygen species; SNHL, sensorineural hearing loss;
SRNS, steroid-resistant nephrotic syndrome; VA, vanillic acid.
References
1 Cain, J.C. and Morton, R. (1955) Some minor constituents of liver oils. Biochem. J. 60, 274–283, https://doi.org/10.1042/bj0600274
2 Crane, F.L., Hatefi, Y., Lester, R. and Widmer, C. (1957) Isolation of a quinone from beed heart mitochondria. Biochim. Biophys. Acta 25, 220–221,
https://doi.org/10.1016/0006-3002(57)90457-2
3 Kaurola,P.,Sharma,V.,Vonk,A.,Vattulainen,I.andR
´
og, T. (2016) Distribution and dynamics of quinones in the lipid bilayer mimicking the inner
membrane of mitochondria. Biochim. Biophys. Acta 1858, 2116–2122, https://doi.org/10.1016/j.bbamem.2016.06.016
4 Evans, D.R. and Guy, H.I. (2004) Mammalian pyrimidine biosynthesis: fresh insights into an ancient pathway. J. Biol. Chem. 279, 33035–33038,
https://doi.org/10.1074/jbc.R400007200
5 Rauchov ´
a, H., Battino, M., Fato, R., Lenaz, G. and Drahota, Z. (1992) Coenzyme Q-pool function in glycerol-3-phosphate oxidation in hamster brown
adipose tissue mitochondria. J. Bioenerg. Biomembr. 24, 235–241, https://doi.org/10.1007/BF00762682
6Mr
´
aˇ
cek, T., Drahota, Z. and Hou ˇ
st ˇ
ek, J. (2013) The function and the role of the mitochondrial glycerol-3-phosphate dehydrogenase in mammalian
tissues. Biochim. Biophys. Acta 1827, 401–410, https://doi.org/10.1016/j.bbabio.2012.11.014
7 Watmough, N.J. and Frerman, F.E. (2010) The electron transfer flavoprotein: ubiquinone oxidoreductases. Biochim. Biophys. Acta 1797, 1910–1916,
https://doi.org/10.1016/j.bbabio.2010.10.007
8 Blake, R.L., Hall, J.G. and Russell, E.S. (1976) Mitochondrial proline dehydrogenase deficiency in hyperprolinemic PRO / re mice: genetic and
enzymatic analyses. Biochem. Genet. 14, 739–757, https://doi.org/10.1007/BF00485338
9 Summit, C.B., Johnson, L.C., J ¨
onsson, T.J., Paesonage, D., Holmes, R.P. and Lowtheir, Q.T. (2015) Proline dehydrogenase 2 (PRODH2) is a
hydroxyproline dehydrogenase (HYPDH) and molecular target for treating primary hyperoxaluria. Biochem. J. 466, 273–281,
https://doi.org/10.1042/BJ20141159
10 Hackfort, B.T. and Mishra, P.K. (2016) Emerging role of hydrogen sulfide-microRNA cross-talk in cardiovascular diseases. Am. J. Physiol. Heart Circ.
Physiol. 310, H802–H812, https://doi.org/10.1152/ajpheart.00660.2015
11 Ziosi, M., Di Meo, I., Kleiner, G., Gao, X.-H., Barca, E., Sanchez-Quintero, M.J. et al. (2017) Coenzyme Q deficiency causes impairment of the sulfide
oxidation pathway. EMBO Mol. Med. 9, 96–111, https://doi.org/10.15252/emmm.201606356
12 Forsmark-Andr ´
ee, P., Dallner, G. and Ernster, L. (1995) Endogenous ubiquinol prevents protein modification accompanying lipid peroxidation in beef
heart submitochondrial particles. Free Radic. Biol. Med. 19, 749–757, https://doi.org/10.1016/0891-5849(95)00076-A
13 Godic, A., Poljsak, B., Adamic, M. and Dahmane, R. (2014) The role of antioxidants in skin cancer prevention and treatment. Oxid. Med. Cell Longev.
2014, 860479, https://doi.org/10.1155/2014/860479
14 Maroz, A., Anderson, R.F., Smith, R.A.J and Murphy, M.P. (2009) Reactivity of ubiquinone and ubiquinol with superoxide and the hydroperoxyl radical:
implications for in vivo antioxidant activity. Free Radic. Biol. Med. 46, 105–109, https://doi.org/10.1016/j.freeradbiomed.2008.09.033
15 Bentinger, M., Brismar, K. and Dallner, G. (2007) The antioxidant role of coenzyme Q. Mitochondrion 7, S41–S50,
https://doi.org/10.1016/j.mito.2007.02.006
16 Mukai, K. (2001) Free radical chemistry of coenzyme Q. In Coenzyme Q Molecular Mechanisms in Health and Disease, 1st edn, Taylor and Francis
Group, pp. 43–61
17 Sohal, R.S. and Forster, M.J. (2007) Coenzyme Q, oxidative stress and aging. Mitochondrion 7, S103–S111,
https://doi.org/10.1016/j.mito.2007.03.006
18 Parrado, C., L ´
opez-Lluch, G., Rodr´
ıguez-Bies, E., Santa-Cruz, S., Navas, P., Ramsey, J. et al. (2011) Calorie restriction modifies ubiquinone and COQ
transcripts levels in mouse tissues. Free Radic. Biol. Med. 50, 1728–1736, https://doi.org/10.1016/j.freeradbiomed.2011.03.024
19 Lopez-Lluch, G., Rodr´
ıguez-Aguilera, J.C., Santos-Oca ˜
na, C. and Navas, P. (2010) Is Coenzyme Q a key factor in aging? Mech. Ageing Dev. 131,
225–235, https://doi.org/10.1016/j.mad.2010.02.003
20 Varela-L ´
opez, A., Giampieri, F., Battino, M. and Quiles, J.L. (2016) Coenzyme Q and its role in the dietary therapy against aging. Molecules 21, 373,
https://doi.org/10.3390/molecules21030373
21 Fernandez-Ayala, D.J., Brea-Calvo, G., Lopez-Lluch, G. and Navas, P. (2005) Coenzyme Q distribution in HL-60 human cells depends on the
endomembrane system. Biochim. Biophys. Acta 1713, 129–137, https://doi.org/10.1016/j.bbamem.2005.05.010
22 Awad, A.M., Bradley, M.C., Fernandez-del-Rio, L., Nag, A., Tsui, H.S. and Clarke, C.F. (2018) Coenzyme Q10 deficiencies: pathways in yeast and
humans. Essays Biochem. 62, 361–376, https://doi.org/10.1042/EBC20170106
c
2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society 393
Essays in Biochemistry (2018) 62 377–398
https://doi.org/10.1042/EBC20170107
23 Payet, L.-A., Leroux, M., Willison, J.C., Kihara, A., Pelosi, L. and Pierrel, F. (2016) Mechanistic details of early steps in coenzyme Q biosynthesis
pathway in yeast. Cell Chem. Biol. 23, 1241–1250, https://doi.org/10.1016/j.chembiol.2016.08.008
24 Stefely, J., Kwiecien, N., Freiberger, E., Richards, A., Jochem, A., Rush, M. et al. (2016) Mitochondrial protein functions elucidated by multi-omic mass
spectrometry profiling. Nat. Biotechnol. 34, 1191–1197, https://doi.org/10.1038/nbt.3683
25 Marbois, B., Xie, L.X., Choi, S., Hirano, K., Hyman, K. and Clarke, C.F. (2010) para-aminobenzoic acid is a precursor in coenzyme Q6 biosynthesis in
Saccharomyces cerevisiae. J. Biol. Chem. 285, 27827–27838, https://doi.org/10.1074/jbc.M110.151894
26 Ozeir, M., M¨
uhlenhoff, U., Webert, H., Lill, R., Fontecave, M. and Pierrel, F. (2011) Coenzyme Q biosynthesis: Coq6 is required for the C5-hydroxylation
reaction and substrate analogs rescue Coq6 deficiency. Chem. Biol. 18, 1134–1142, https://doi.org/10.1016/j.chembiol.2011.07.008
27 Jonassen, T. and Clarke, C.F. (2000) Isolation and functional expression of human COQ3, a gene encoding a methyltransferase required for ubiquinone
biosynthesis. J. Biol. Chem. 275, 12381–12387, https://doi.org/10.1074/jbc.275.17.12381
28 Poon, W.W., Barkovich, R.J., Hsu, Y., Frankel, A., Lee, P.T., Shepherd, N. et al. (1999) Yeast and Rat Coq3 and Escherichia coli UbiG polypeptides
catalyze both O -methyltransferase steps in coenzyme Q biosynthesis. J. Biol. Chem. 274, 21665–21672, https://doi.org/10.1074/jbc.274.31.21665
29 Barkovich, R.J., Shtanko, A., Shepherd, J.A., Lee, P.T., Myles, D.C., Tzagoloff, A. et al. (1997) Characterization of the COQ5 Gene from Saccharomyces
cerevisiae. Biochemistry 272, 9182–9188
30 Nguyen, T.P.T, Casarin, A., Desbats, M.A., Doimo, M., Trevisson, E., Santos-Oca˜
na, C. et al. (2014) Molecular characterization of the human COQ5
C-methyltransferase in Coenzyme Q10 biosynthesis. Biochim. Biophys. Acta 1841, 1628–1638, https://doi.org/10.1016/j.bbalip.2014.08.007
31 Marbois, B.N. and Clarke, C.F. (1996) The COQ7 gene encodes a protein in saccharomyces cerevisiae necessary for ubiquinone biosynthesis. J. Biol.
Chem. 271, 2995–3004, https://doi.org/10.1074/jbc.271.6.2995
32 Pierrel, F., Hamelin, O., Douki, T., Kieffer-Jaquinod, S., M¨
uhlenhoff, U., Ozeir, M. et al. (2010) Involvement of mitochondrial ferredoxin and
para-aminobenzoic acid in yeast coenzyme Q biosynthesis. Chem. Biol. 17, 449–459, https://doi.org/10.1016/j.chembiol.2010.03.014
33 Tauche, A., Krause-Buchholz, U. and R¨
odel, G. (2008) Ubiquinone biosynthesis in Saccharomyces cerevisiae: The molecular organization of
O-methylase Coq3p depends on Abc1p/Coq8p. FEMS Yeast Res. 8, 1263–1275, https://doi.org/10.1111/j.1567-1364.2008.00436.x
34 Xie, L.X., Hsieh, E.J., Watanabe, S., Allan, C.M., Chen, J.Y., Tran, U.C. et al. (2011) Expression of the human atypical kinase ADCK3 rescues coenzyme
Q biosynthesis and phosphorylation of Coq polypeptides in yeast coq8 mutants. Biochim. Biophys. Acta 1811, 348–360,
https://doi.org/10.1016/j.bbalip.2011.01.009
35 Stefely, J.A., Reidenbach, A.G., Ulbrich, A., Oruganty, K., Floyd, B.J., Jochem, A. et al. (2015) Mitochondrial ADCK3 employs an atypical protein
kinase-like fold to enable coenzyme Q biosynthesis. Mol. Cell 57, 83–94, https://doi.org/10.1016/j.molcel.2014.11.002
36 Reidenbach, A.G., Kemmerer, Z.A., Aydin, D., Jochem, A., McDevitt, M.T., Hutchins, P.D. et al. (2018) Conserved lipid and small-molecule modulation
of COQ8 reveals regulation of the ancient kinase-like UbiB family. Cell Chem. Biol. 25, 154.e11–165.e11,
https://doi.org/10.1016/j.chembiol.2017.11.001
37 Marbois, B., Gin, P., Gulmezian, M. and Clarke, C.F. (2009) The yeast Coq4 polypeptide organizes a mitochondrial protein complex essential for
Coenzyme Q biosynthesis. Biochim. Biophys. Acta 1791, 69–75, https://doi.org/10.1016/j.bbalip.2008.10.006
38 Hsieh, E.J., Gin, P., Gulmezian, M., Tran, U.P.C, Saiki, R., Marbois, B.N. et al. (2007) Saccharomyces cerevisiae Coq9 polypeptide is a subunit of the
mitochondrial Coenzyme Q biosynthetic complex. Arch. Biochem. Biophys. 463, 19–26, https://doi.org/10.1016/j.abb.2007.02.016
39 Lohman, D.C., Forouhar, F., Beebe, E.T., Stefely, M.S., Minogue, C.E., Ulbrich, A. et al. (2014) Mitochondrial COQ9 is a lipid-binding protein that
associates with COQ7 to enable Coenzyme Q biosynthesis. Proc.Natl.Acad.Sci.U.S.A.111, E4697–e4705,
https://doi.org/10.1073/pnas.1413128111
40 Cui, T.Z. and Kawamukai, M. (2009) Coq10, a mitochondrial Coenzyme Q binding protein, is required for proper respiration in Schizosaccharomyces
pombe. FEBS J. 276, 748–759, https://doi.org/10.1111/j.1742-4658.2008.06821.x
41 Allan, C.M., Awad, A.M., Johnson, J.S., Shirasaki, D.I., Wang, C., Blaby-Haas, C.E. et al. (2015) Identification of Coq11, a new coenzyme Q
biosynthetic protein in the CoQ-synthome in saccharomyces cerevisiae. J. Biol. Chem. 290, 7517–7534, https://doi.org/10.1074/jbc.M114.633131
42 Desbats, M.A., Lunardi, G., Doimo, M., Trevisson, E. and Salviati, L. (2015) Genetic bases and clinical manifestations of coenzyme Q10 (CoQ 10)
deficiency. J. Inherit. Metab. Dis. 38, 145–156
43 Padilla, S., Tran, U.C., Jim´
enez-Hidalgo, M., L ´
opez-Mart´
ın, J.M., Mart´
ın-Montalvo, A., Clarke, C.F. et al. (2009) Hydroxylation of demethoxy-Q6
constitutes a control point in yeast coenzyme Q6 biosynthesis. Cell. Mol. Life Sci. 66, 173–186, https://doi.org/10.1007/s00018-008-8547-7
44 He, C.H., Xie, L.X., Allan, C.M., Tran, U.C. and Clarke, C.F. (2014) Coenzyme Q supplementation or over-expression of the yeast Coq8 putative kinase
stabilizes multi-subunit Coq polypeptide complexes in yeast coq null mutants. Biochim. Biophys. Acta 1841, 630–644,
https://doi.org/10.1016/j.bbalip.2013.12.017
45 Poon, W.W., Do, T.Q., Noelle Marbois, B. and Clarke, C.F. (1997) Sensitivity to treatment with polyunsaturated fatty acids is a general characteristic of
the ubiquinone-deficient yeast coq mutants. Mol. Aspects Med. 18, 121–127, https://doi.org/10.1016/S0098-2997(97)00004-6
46 Baba, S.W., Belogrudov, G.I., Lee, J.C., Lee, P.T., Strahan, J., Shepherd, J.N. et al. (2004) Yeast Coq5 C-methyltransferase is required for stability of
other polypeptides involved in Coenzyme Q biosynthesis. J. Biol. Chem. 279, 10052–10059, https://doi.org/10.1074/jbc.M313712200
47 Gin, P. and Clarke, C.F. (2005) Genetic evidence for a multi-subunit complex in coenzyme Q Biosynthesis in yeast and the role of the Coq1 hexaprenyl
diphosphate synthase. J. Biol. Chem. 280, 2676–2681, https://doi.org/10.1074/jbc.M411527200
48 Gonzalez-Mariscal, I., Garcia-Teston, E., Padilla, S., Martin-Montalvo, A., Pomares-Viciana, T., Vazquez-Fonseca, L. et al. (2014) Regulationof
coenzyme Q biosynthesis in yeast: a new complex in the block. IUBMB Life 66, 63–70, https://doi.org/10.1002/iub.1243
49 Stefely, J.A., Licitra, F., Laredj, L., Reidenbach, A.G., Kemmerer, Z.A., Grangeray, A. et al. (2016) Cerebellar ataxia and coenzyme Q deficiency through
loss of unorthodox kinase activity. Mol. Cell 63, 608–620, https://doi.org/10.1016/j.molcel.2016.06.030
50 Luna-Sanchez, M., Diaz-Casado, E., Barca, E., Tejada, M.A., Montilla-Garcia, A., Cobos, E.J. et al. (2015) The clinical heterogeneity of CoenzymeQ10
deficiency results from genotypic differences in the Coq9 gene. EMBO Mol. Med. 7, 670–687, https://doi.org/10.15252/emmm.201404632
394 c
2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society
Essays in Biochemistry (2018) 62 377–398
https://doi.org/10.1042/EBC20170107
51 Ashraf, S., Gee, H.Y., Woerner, S., Xie, L.X., Vega-Warner, V., Lovric, S. et al. (2013) ADCK4 mutations promote steroid-resistant nephrotic syndrome
through CoQ10 biosynthesis disruption. J. Clin. Invest. 123, 5179–5189, https://doi.org/10.1172/JCI69000
52 Floyd, B.J., Wilkerson, E.M., Veling, M.T., Minogue, C.E., Xia, C., Beebe, E.T. et al. (2016) Mitochondrial protein interaction mapping identifies
regulators of respiratory chain function. Mol. Cell 63, 621–632, https://doi.org/10.1016/j.molcel.2016.06.033
53 Yen, H.C., Liu, Y.C., Kan, C.C., Wei, H.J., Lee, S.H., Wei, Y.H. et al. (2016) Disruption of the human COQ5-containing protein complex is associated with
diminished Coenzyme Q10 levels under two different conditions of mitochondrial energy deficiency. Biochim. Biophys. Acta 1860, 1864–1876,
https://doi.org/10.1016/j.bbagen.2016.05.005
54 Turunen, M., Olsson, J. and Dallner, G. (2004) Metabolism and function of Coenzyme Q. Biochim. Biophys. Acta 1660, 171–199,
https://doi.org/10.1016/j.bbamem.2003.11.012
55 Bentinger, M., Turunen, M., Zhang, X.-X., Yvonne Wan, Y.-J. and Dallner, G. (2003) Involvement of retinoid X receptor αin coenzyme Q metabolism. J.
Mol. Biol. 326, 795–803, https://doi.org/10.1016/S0022-2836(02)01447-X
56 Brea-Calvo, G., Siendones, E., S ´
anchez-Alc ´
azar, J.A., de Cabo, R. and Navas, P. (2009) Cell survival from chemotherapy depends on NF-kappaB
transcriptional up-regulation of Coenzyme Q biosynthesis. PLoS One 4, e5301, https://doi.org/10.1371/journal.pone.0005301
57 Cascajo, M.V., Abdelmohsen, K., Heon Noh, J., Fernandez-Ayala, D.J., Willers, I.M., Brea, G. et al. (2016) RNA-binding proteins regulate cell respiration
and coenzyme Q biosynthesis by post-transcriptional regulation of COQ7. RNA Biol 13, 622–634, https://doi.org/10.1080/15476286.2015.1119366
58 Lapointe, C.P., Stefely, J.A., Jochem, A., Hutchins, P.D., Wilson, G.M., Kwiecien, N.W. et al. (2018) Multi-omics reveal specific targets of the
RNA-Binding protein puf3p and its orchestration of mitochondrial biogenesis. Cell Systems 125.e6–135.e6
59 Veling, M.T., Reidenbach, A.G., Freiberger, E.C., Kwiecien, N.W., Hutchins, P.D., Drahnak, M.J. et al. (2017) Multi-omic mitoprotease profiling defines a
role for Oct1p in Coenzyme Q production. Mol. Cell 68, 970.e11–977.e11, https://doi.org/10.1016/j.molcel.2017.11.023
60 Mart´
ın-Montalvo, A., Gonz´
alez-Mariscal, I., Pomares-Viciana, T., Padilla-L´
opez, S., Ballesteros, M., Vazquez-Fonseca, L. et al. (2013) The phosphatase
Ptc7 induces Coenzyme Q biosynthesis by activating the hydroxylase Coq7 in yeast. J. Biol. Chem. 288, 28126–28137,
https://doi.org/10.1074/jbc.M113.474494
61 Vasta, V., Merritt, II, J.L., Saneto, R.P. and Hahn, S.H. (2012) Next-generation sequencing for mitochondrial diseases: a wide diagnostic spectrum.
Pediatr. Int. 54, 585–601, https://doi.org/10.1111/j.1442-200X.2012.03644.x
62 Mollet, J., Giurgea, I., Schlemmer, D., Dallner, G., Chretien, D., Delahodde, A. et al. (2007) Prenyldiphosphate synthase, subunit 1 (PDSS1) and
OH-benzoate polyprenyltransferase (COQ2) mutations in ubiquinone deficiency and oxidative phosphorylation disorders. J. Clin. Invest. 117, 765–772,
https://doi.org/10.1172/JCI29089
63 Iv ´
anyi, B., R´
acz, G.Z., G´
al, P., Brinyiczki, K., B´
odi, I., Kalm´
ar, T. et al. (2018) Diffuse mesangial sclerosis in a PDSS2 mutation-induced coenzyme Q10
deficiency. Pediatr. Nephrol. 33, 439–446, https://doi.org/10.1007/s00467-017-3814-1
64 L ´
opez, L.C., Schuelke, M., Quinzii, C.M., Kanki, T., Rodenburg, R.J.T, Naini, A. et al. (2006) Leigh syndrome with nephropathy and CoQ10 deficiency
due to decaprenyl diphosphate synthase subunit 2 (PDSS2) mutations. Am.J.Hum.Genet.79, 1125–1129, https://doi.org/10.1086/510023
65 Sadowski, C.E., Lovric, S., Ashraf, S., Pabst, W.L., Gee, H.Y., Kohl, S. et al. (2015) A single-gene cause in 29.5% of cases of steroid-resistant nephrotic
syndrome. J. Am. Soc. Nephrol. 26, 1279–1289, https://doi.org/10.1681/ASN.2014050489
66 R ¨
otig, A., Appelkvist, E.L., Geromel, V., Chretien, D., Kadhom, N., Edery, P. et al. (2000) Quinone-responsive multiple respiratory-chain dysfunction due
to widespread coenzyme Q10deficiency. Lancet 356, 391–395, https://doi.org/10.1016/S0140-6736(00)02531-9
67 Rahman, S., Clarke, C.F. and Hirano, M. (2012) 176th ENMC International Workshop: diagnosis and treatment of coenzyme Q10 deficiency.
Neuromuscul. Disord. 22, 76–86, https://doi.org/10.1016/j.nmd.2011.05.001
68 Starr, M.C., Chang, I.J., Finn, L.S., Sun, A., Larson, A.A., Goebel, J. et al. (2018) COQ2 nephropathy: a treatable cause of nephrotic syndrome in
children. Pediatr. Nephrol. 1–5
69 Mitsui, J., Matsukawa, T., Ishiura, H., Fukuda, Y., Ichikawa, Y., Date, H. et al. (2013) Mutations in COQ2 in familial and sporadic multiple-system
atrophy. N. Engl. J. Med. 369, 233–244, https://doi.org/10.1056/NEJMoa1212115
70 Desbats, M.A., Morbidoni, V., Silic-Benussi, M., Doimo, M., Ciminale, V., Cassina, M. et al. (2016) The COQ2 genotype predicts the severity of
Coenzyme Q10 deficiency. Hum. Mol. Genet. 25, 4256–4265, https://doi.org/10.1093/hmg/ddw257
71 Scalais, E., Chafai, R., Van Coster, R., Bindl, L., Nuttin, C., Panagiotaraki, C. et al. (2013) Early myoclonic epilepsy, hypertrophic cardiomyopathy and
subsequently a nephrotic syndrome in a patient with CoQ10 deficiency caused by mutations in para-hydroxybenzoate-polyprenyl transferase (COQ2).
Eur. J. Paediatr. Neurol. 17, 625–630, https://doi.org/10.1016/j.ejpn.2013.05.013
72 Diomedi-Camassei, F., Di Giandomenico, S., Santorelli, F.M., Caridi, G., Piemonte, F., Montini, G. et al. (2007) COQ2 nephropathy: a newly described
inherited mitochondriopathy with primary renal involvement. J. Am. Soc. Nephrol. 18, 2773–2780, https://doi.org/10.1681/ASN.2006080833
73 Dinwiddie, D.L., Smith, L.D., Miller, N.A., Atherton, A.M., Farrow, E.G., Strenk, M.E. et al. (2013) Diagnosis of mitochondrial disorders by concomitant
next-generation sequencing of the exome and mitochondrial genome. Genomics 102, 148–156, https://doi.org/10.1016/j.ygeno.2013.04.013
74 Desbats, M.A., Vetro, A., Limongelli, I., Lunardi, G., Casarin, A., Doimo, M. et al. (2015) Primary coenzyme Q 10 deficiency presenting as fatal neonatal
multiorgan failure. Eur. J. Hum. Genet. 23, 1254–1258, https://doi.org/10.1038/ejhg.2014.277
75 McCarthy, H.J., Bierzynska, A., Wherlock, M., Ognjanovic, M., Kerecuk, L., Hegde, S. et al. (2013) Simultaneous sequencing of 24 genes associated
with steroid-resistant nephrotic syndrome. Clin. J. Am. Soc. Nephrol. 8, 637–648, https://doi.org/10.2215/CJN.07200712
76 Salviati, L., Sacconi, S., Murer, L., Zacchello, G., Franceschini, L., Laverda, A.M. et al. (2005) Infantile encephalomyopathy and nephropathy with
CoQ10 deficiency: a CoQ10-responsive condition. Neurology 65, 606–608, https://doi.org/10.1212/01.wnl.0000172859.55579.a7
77 Quinzii, C., Naini, A., Salviati, L., Trevisson, E., Navas, P., Dimauro, S. et al. (2006) A mutation in para-hydroxybenzoate-polyprenyl transferase (COQ2)
causes primary coenzyme Q10 deficiency. Am.J.Hum.Genet.78, 345–349, https://doi.org/10.1086/500092
78 Jakobs, B.S., van den Heuvel, L.P., Smeets, R.J.P, de Vries, M.C., Hien, S., Schaible, T. et al. (2013) A novel mutation in COQ2 leading to fatal infantile
multisystem disease. J. Neurol. Sci. 326, 24–28, https://doi.org/10.1016/j.jns.2013.01.004
c
2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society 395
Essays in Biochemistry (2018) 62 377–398
https://doi.org/10.1042/EBC20170107
79 Gigante, M., Diella, S., Santangelo, L., Trevisson, E., Acosta, M.J., Amatruda, M. et al. (2017) Further phenotypic heterogeneity of CoQ10 deficiency
associated with steroid resistant nephrotic syndrome and novel COQ2 and COQ6 variants. Clin. Genet. 92, 224–226,
https://doi.org/10.1111/cge.12960
80 Sondheimer, N., Hewson, S., Cameron, J.M., Somers, G.R., Broadbent, J.D., Ziosi, M. et al. (2017) Novel recessive mutations in COQ4 cause severe
infantile cardiomyopathy and encephalopathy associated with CoQ 10 deficiency. Mol. Genet. Metab. Rep. 12, 23–27,
https://doi.org/10.1016/j.ymgmr.2017.05.001
81 Brea-Calvo, G., Haack, T.B., Karall, D., Ohtake, A., Invernizzi, F., Carrozzo, R. et al. (2015) COQ4 mutations cause a broad spectrum of mitochondrial
disorders associated with CoQ10 deficiency. Am.J.Hum.Genet.96, 309–317, https://doi.org/10.1016/j.ajhg.2014.12.023
82 Chung, W.K., Martin, K., Jalas, C., Braddock, S.R., Juusola, J., Monaghan, K.G. et al. (2015) Mutations in COQ4, an essential component of coenzyme
Q biosynthesis, cause lethal neonatal mitochondrial encephalomyopathy. J. Med. Genet. 52, 627–635,
https://doi.org/10.1136/jmedgenet-2015-103140
83 Helbig, K.L., Farwell Hagman, K.D., Shinde, D.N., Mroske, C., Powis, Z., Li, S. et al. (2016) Diagnostic exome sequencing provides a molecular
diagnosis for a significant proportion of patients with epilepsy. Genet. Med. 18, 898–905, https://doi.org/10.1038/gim.2015.186
84 Salviati, L., Trevisson, E., Rodriguez Hernandez, M.A., Casarin, A., Pertegato, V., Doimo, M. et al. (2012) Haploinsufficiency of COQ4 causes coenzyme
Q10 deficiency. J. Med. Genet. 49, 187–191, https://doi.org/10.1136/jmedgenet-2011-100394
85 Malicdan, M.C.V., Vilboux, T., Ben-Zeev, B., Guo, J., Eliyahu, A., Pode-Shakked, B. et al. (2018) A novel inborn error of the coenzyme Q10 biosynthesis
pathway: cerebellar ataxia and static encephalomyopathy due to COQ5 C-methyltransferase deficiency. Hum. Mutat. 39, 69–79,
https://doi.org/10.1002/humu.23345
86 Park, E., Ahn, Y.H., Kang, H.G., Yoo, K.H., Won, N.H., Lee, K.B. et al. (2017) COQ6 mutations in children with steroid-resistant focal segmental
glomerulosclerosis and sensorineural hearing loss. Am.J.KidneyDis.70, 139–144, https://doi.org/10.1053/j.ajkd.2016.10.040
87 Heeringa, S.F., Chernin, G., Chaki, M., Zhou, W., Sloan, A.J., Ji, Z. et al. (2011) COQ6 mutations in human patients produce nephrotic syndrome with
sensorineural deafness. J. Clin. Invest. 121, 2013–2024, https://doi.org/10.1172/JCI45693
88 Doimo, M., Trevisson, E., Airik, R., Bergdoll, M., Santos-Oca˜
na, C., Hildebrandt, F. et al. (2014) Effect of vanillic acid on COQ6 mutants identified in
patients with coenzyme Q10deficiency. Biochim. Biophys. Acta 1842,16, https://doi.org/10.1016/j.bbadis.2013.10.007
89 Cao, Q., Li, G.M., Xu, H., Shen, Q., Sun, L., Fang, X.Y. et al. (2017) Coenzyme Q(10) treatment for one child with COQ6 gene mutation induced
nephrotic syndrome and literature review. Zhonghua er ke za zhi Chinese J. Pediatr. 55, 135–138
90 Wang, Y., Smith, C., Parboosingh, J.S., Khan, A., Innes, M. and Hekimi, S. (2017) Pathogenicity of two COQ7 mutations and responses to
2,4-dihydroxybenzoate bypass treatment. J. Cell. Mol. Med. 21, 2329–2343, https://doi.org/10.1111/jcmm.13154
91 Freyer, C., Stranneheim, H., Naess, K., Mourier, A., Felser, A., Maffezzini, C. et al. (2015) Rescue of primary ubiquinone deficiency due to a novel COQ7
defect using 2,4-dihydroxybensoic acid. J. Med. Genet. 52, 779–783, https://doi.org/10.1136/jmedgenet-2015-102986
92 Danhauser, K., Herebian, D., Haack, T.B., Rodenburg, R.J., Strom, T.M., Meitinger, T. et al. (2016) Fatal neonatal encephalopathy and lactic acidosis
caused by a homozygous loss-of-function variant in COQ9. Eur. J. Hum. Genet. 24, 450–454, https://doi.org/10.1038/ejhg.2015.133
93 Smith, A.C., Ito, Y., Ahmed, A., Schwartzentruber, J.A., Beaulieu, C.L., Aberg, E. et al. (2018) A family segregating lethal neonatal coenzyme Q 10
deficiency caused by mutations in COQ9. J. Inherit. Metab. Dis.,https://doi.org/10.1007/s10545-017-0122-7
94 Rahman, S., Hargreaves, I., Clayton, P. and Heales, S. (2001) Neonatal presentation of coenzyme Q10 deficiency. J. Pediatr. 139, 456–458,
https://doi.org/10.1067/mpd.2001.117575
95 Duncan, A.J., Bitner-Glindzicz, M., Meunier, B., Costello, H., Hargreaves, I.P., L´
opez, L.C. et al. (2009) A nonsense mutation in COQ9 causes
autosomal-recessive neonatal-onset primary coenzyme Q10 deficiency: a potentially treatable form of mitochondrial disease. Am.J.Hum.Genet.84,
558–566, https://doi.org/10.1016/j.ajhg.2009.03.018
96 Lagier-Tourenne, C., Tazir, M., L´
opez, L.C., Quinzii, C.M., Assoum, M., Drouot, N. et al. (2008) ADCK3, an ancestral kinase, is mutated in a form of
recessive ataxia associated with coenzyme Q10 deficiency. Am.J.Hum.Genet.82, 661–672, https://doi.org/10.1016/j.ajhg.2007.12.024
97 Mignot, C., Apartis, E., Durr, A., Marques Lourenco, C., Charles, P., Devos, D. et al. (2013) Phenotypic variability in ARCA2 and identification of a core
ataxic phenotype with slow progression. Orphanet J. Rare Dis. 8, 173, https://doi.org/10.1186/1750-1172-8-173
98 Mollet, J., Delahodde, A., Serre, V., Chretien, D., Schlemmer, D., Lombes, A. et al. (2008) CABC1 gene mutations cause ubiquinone deficiency with
cerebellar ataxia and seizures. Am.J.Hum.Genet.82, 623–630, https://doi.org/10.1016/j.ajhg.2007.12.022
99 Horvath, R., Czermin, B., Gulati, S., Demuth, S., Houge, G., Pyle, A. et al. (2012) Adult-onset cerebellar ataxia due to mutations in CABC1/ADCK3. J.
Neurol. Neurosurg. Psychiatry 83, 174–178, https://doi.org/10.1136/jnnp-2011-301258
100 Pronicka, E., Piekutowska-Abramczuk, D., Ciara, E., Trubicka, J., Rokicki, D., Karkucinska-Wieckowska, A. et al. (2016) New perspective in
diagnostics of mitochondrial disorders: two years’ experience with whole-exome sequencing at a national paediatric centre. J. Transl. Med. 14, 174,
https://doi.org/10.1186/s12967-016-0930-9
101 Jacobsen, J.C., Whitford, W., Swan, B., Taylor, J., Love, D.R., Hil, R. et al. (2017) Compound heterozygous inheritance of mutations in coenzyme Q8A
results in autosomal recessive cerebellar ataxia and coenzyme Q10 deficiency in a female sib-pair. JIMD Rep. 1–6,
https://doi.org/10.1007/8904˙2017˙73
102 Hikmat, O., Tzoulis, C., Knappskog, P.M., Johansson, S., Boman, H., Sztromwasser, P. et al. (2016) ADCK3 mutations with epilepsy, stroke-like
episodes and ataxia: a POLG mimic? Eur. J. Neurol. 23, 1188–1194, https://doi.org/10.1111/ene.13003
103 Anheim, M., Fleury, M., Monga, B., Laugel, V., Chaigne, D., Rodier, G. et al. (2010) Epidemiological, clinical, paraclinical and molecular study of a
cohort of 102 patients affected with autosomal recessive progressive cerebellar ataxia from Alsace, Eastern France: Implications for clinical
management. Neurogenetics 11, 1–12, https://doi.org/10.1007/s10048-009-0196-y
104 Gerards, M., van den Bosch, B., Calis, C., Schoonderwoerd, K., van Engelen, K., Tijssen, M. et al. (2010) Nonsense mutations in CABC1/ADCK3 cause
progressive cerebellar ataxia and atrophy. Mitochondrion 10, 510–515, https://doi.org/10.1016/j.mito.2010.05.008
396 c
2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society
Essays in Biochemistry (2018) 62 377–398
https://doi.org/10.1042/EBC20170107
105 Terracciano, A., Renaldo, F., Zanni, G., D’Amico, A., Pastore, A., Barresi, S. et al. (2012) The use of muscle biopsy in the diagnosis of undefined ataxia
with cerebellar atrophy in children. Eur. J. Paediatr. Neurol. 16, 248–256, https://doi.org/10.1016/j.ejpn.2011.07.016
106 Barca, E., Musumeci, O., Montagnese, F., Marino, S., Granata, F., Nunnari, D. et al. (2016) Cerebellar ataxia and severe muscle CoQ 10 deficiency in a
patient with a novel mutation in ADCK3. Clin. Genet. 90, 156–160, https://doi.org/10.1111/cge.12742
107 Blumkin, L., Leshinsky-Silver, E., Zerem, A., Yosovich, K., Lerman-Sagie, T. and Lev, D. (2014) Heterozygous mutations in the ADCK3 gene in siblings
with cerebellar atrophy and extreme phenotypic variability. JIMD Rep. 12, 103–107, https://doi.org/10.1007/8904˙2013˙251
108 Liu, Y.T., Hersheson, J., Plagnol, V., Fawcett, K., Duberley, K.E.C, Preza, E. et al. (2014) Autosomal-recessive cerebellar ataxia caused by a novel
ADCK3 mutation that elongates the protein:Clinical, genetic and biochemical characterisation. J. Neurol. Neurosurg. Psychiatry 85, 493–498,
https://doi.org/10.1136/jnnp-2013-306483
109 Malgireddy, K., Thompson, R. and Torres-Russotto, D. (2016) A novel CABC1/ADCK3 mutation in adult-onset cerebellar ataxia. Park Relat. Disord. 33,
151–152, https://doi.org/10.1016/j.parkreldis.2016.10.010
110 Atmaca, M., Gulhan, B., Korkmaz, E., Inozu, M., Soylemezoglu, O., Candan, C. et al. (2017) Follow-up results of patients with ADCK4 mutations and
the efficacy of CoQ10 treatment. Pediatr. Nephrol. 32, 1369–1375, https://doi.org/10.1007/s00467-017-3634-3
111 Korkmaz, E., Lipska-Zietkiewicz, B.S., Boyer, O., Gribouval, O., Fourrage, C., Tabatabaei, M. et al. (2016) ADCK4-associated glomerulopathy causes
adolescence-onset FSGS. J. Am. Soc. Nephrol. 27, 63–68, https://doi.org/10.1681/ASN.2014121240
112 Park, E., Kang, H.G., Choi, Y.H., Lee, K.B., Moon, K.C., Jeong, H.J. et al. (2017) Focal segmental glomerulosclerosis and medullary nephrocalcinosis in
children with ADCK4 mutations. Pediatr. Nephrol. 32, 1547–1554, https://doi.org/10.1007/s00467-017-3657-9
113 Vazquez-Fonseca, L., Doimo, M., Calderan, C., Desbats, M.A., Acosta, M.J., Cerqua, C. et al. (2018) Mutations in COQ8B (ADCK4) found in patients
with steroid resistant nephrotic syndrome alter COQ8B function. Hum. Mutat. 39, 406–414, https://doi.org/10.1002/humu.23376
114 Feng, C., Wang, Q., Wang, J., Liu, F., Shen, H., Fu, H. et al. (2017) Coenzyme Q10 supplementation therapy for 2 children with proteinuria renal
disease and ADCK4 mutation. Medicine 96, e8880
115 Quinzii, C.M., Lopez, L.C., Gilkerson, R.W., Dorado, B., Coku, J., Naini, A.B. et al. (2010) Reactive oxygen species, oxidative stress, and cell death
correlate with level of CoQ10 deficiency. FASEB J. 24, 3733–3743, https://doi.org/10.1096/fj.09-152728
116 Lolin, K., Chiodini, B.D., Hennaut, E., Adams, B., Dahan, K. and Ismaili, K. (2017) Early-onset of ADCK4 glomerulopathy with renal failure: a case
report. BMC Med. Genet. 18, 28, https://doi.org/10.1186/s12881-017-0392-9
117 Zhang, H., Wang, F., Liu, X., Zhong, X., Yao, Y. and Xiao, H. (2017) Steroid-resistant nephrotic syndrome caused by co-inheritance of mutations at
NPHS1 and ADCK4 genes in two Chinese siblings. Intractable Rare Dis. Res. 6, 299–303, https://doi.org/10.5582/irdr.2017.01037
118 Hughes, B.G., Harrison, P.M. and Hekimi, S. (2017) Estimating the occurrence of primary ubiquinone deficiency by analysis of large-scale sequencing
data. Sci. Rep. 7, 17744, https://doi.org/10.1038/s41598-017-17564-y
119 Ogaki, K., Fujioka, S., Heckman, M.G., Rayaprolu, S., Soto-Ortolaza, A.I., Labb ´
e, C. et al. (2014) Analysis of COQ2 gene in multiple system atrophy.
Mol. Neurodegener 9, 44, https://doi.org/10.1186/1750-1326-9-44
120 Louw, R., Smuts, I., Wilsenach, K.-L., Jonck, L.-M., Schoonen, M. and van der Westhuizen, F.H. (2018) The dilemma of diagnosing Coenzyme Q 10
deficiency in muscle. Mol. Genet. Metab.,https://doi.org/10.1016/j.ymgme.2018.02.015
121 Emmanuele, V., L ´
opez, L.C., L´
opez, L., Berardo, A., Naini, A., Tadesse, S. et al. (2012) Heterogeneity of coenzyme Q10 deficiency: patient study and
literature review. Arch. Neurol. 69, 978–983, https://doi.org/10.1001/archneurol.2012.206
122 L ´
opez-Mart´
ın, J.M., Salviati, L., Trevisson, E., Montini, G., DiMauro, S., Quinzii, C. et al. (2007) Missense mutation of the COQ2 gene causes defects of
bioenergetics and de novo pyrimidine synthesis. Hum. Mol. Genet. 16, 1091–1097, https://doi.org/10.1093/hmg/ddm058
123 Rodr´
ıguez-Hern ´
andez, ´
A, Cordero, M.D., Salviati, L., Artuch, R., Pineda, M., Briones, P. et al. (2009) Coenzyme Q deficiency triggers mitochondria
degradation by mitophagy. Autophagy 5, 19–32, https://doi.org/10.4161/auto.5.1.7174
124 Peng, M., Ostrovsky, J., Kwon, Y.J., Polyak, E., Licata, J., Tsukikawa, M. et al. (2015) Inhibiting cytosolic translation and autophagy improves health in
mitochondrial disease. Hum. Mol. Genet. 24, 4829–4847, https://doi.org/10.1093/hmg/ddv207
125 Quinzii, C.M., Luna-Sanchez, M., Ziosi, M., Hidalgo-Gutierrez, A., Kleiner, G. and Lopez, L.C. (2017) The role of sulfide oxidation impairment in the
pathogenesis of primary CoQ deficiency. Front. Physiol. 8, 525, https://doi.org/10.3389/fphys.2017.00525
126 Fazakerley, D.J., Chaudhuri, R., Yang, P., Maghzal, G.J., Thomas, K.C., Krycer, J.R. et al. (2018) Mitochondrial CoQ deficiency is a common driver of
mitochondrial oxidants and insulin resistance. Elife 7, e32111, https://doi.org/10.7554/eLife.32111
127 Salviati, L., Trevisson, E., Doimo, M., Navas, P. et al. (2017) Primary Coenzyme Q10 Deficiency. In GeneReviews R
(Adam, M.P., Ardinger, H.H., Pagon,
R.A., Wallace, S.E., Bean, L.J.H and Stephens, K., eds), pp. 1993–2018, University of Washington, Seattle
128 Yubero, D., Montero, R., Amstrong, J., Espinos, C., Palau, F., Santos-Oca ˜
na, C. et al. (2015) Molecular diagnosis of coenzyme Q10 deficiency. Expert
Rev. Mol. Diagn. 15, 1049–1059, https://doi.org/10.1586/14737159.2015.1062727
129 Rodr´
ıguez-Aguilera, J.C., Cort ´
es, A.B., Fern´
andez-Ayala, D.J.M and Navas, P. (2017) Biochemical assessment of coenzyme Q10 deficiency. J. Clin.
Med. 6, E27, https://doi.org/10.3390/jcm6030027
130 Yubero, D., Allen, G., Artuch, R. and Montero, R. (2017) The value of coenzyme q10 determination in mitochondrial patients. J. Clin. Med. 6,37,
https://doi.org/10.3390/jcm6040037
131 Artuch, R., Brea-Calvo, G., Briones, P., Aracil, A., Galv ´
an, M., Espin´
os, C. et al. (2006) Cerebellar ataxia with coenzyme Q10 deficiency: diagnosis and
follow-up after coenzyme Q10 supplementation. J. Neurol. Sci. 246, 153–158, https://doi.org/10.1016/j.jns.2006.01.021
132 Yubero, D., Montero, R., Ramos, M., Neergheen, V., Navas, P., Artuch, R. et al. (2015) Determination of urinary coenzyme Q 10 by HPLC with
electrochemical detection: reference values for a paediatric population. Biofactors 41, 424–430, https://doi.org/10.1002/biof.1242
133 Bhagavan, H.N. and Chopra, R.K. (2006) Coenzyme Q10: absorption, tissue uptake, metabolism and pharmacokinetics. Free Radic. Res. 40, 445–453,
https://doi.org/10.1080/10715760600617843
134 Zaki, N.M. (2016) Strategies for oral delivery and mitochondrial targeting of CoQ10. Drug Deliv. 23, 1868–1881
c
2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society 397
Essays in Biochemistry (2018) 62 377–398
https://doi.org/10.1042/EBC20170107
135 Stocker, R., Bowry, V.W. and Frei, B. (1991) Ubiquinol-10 protects human low density lipoprotein more efficiently against lipid peroxidation than does
alpha-tocopherol. Proc.Natl.Acad.Sci.U.S.A.88, 1646–1650, https://doi.org/10.1073/pnas.88.5.1646
136 Mohr, D., Bowry, V.W. and Stocker, R. (1992) Dietary supplementation With coenzyme Q 10 results in increased levels of ubiquinol-10 within
circulating lipoproteins and increased resistance of human low density lipoprotein to the initiation of lipid peroxidation. Biochim. Biophys. Acta 1126,
247–254, https://doi.org/10.1016/0005-2760(92)90237-P
137 Potgieter, M., Pretorius, E. and Pepper, M.S. (2013) Primary and secondary coenzyme Q10 deficiency: the role of therapeutic supplementation. Nutr.
Rev. 71, 180–188, https://doi.org/10.1111/nure.12011
138 Montini, G., Malaventura, C. and Salviati, L. (2008) Early coenzyme Q10 supplementation in primary coenzyme Q10 deficiency. N. Engl. J. Med. 358,
2849–2850, https://doi.org/10.1056/NEJMc0800582
139 Saiki, R., Lunceford, A.L., Shi, Y., Marbois, B., King, R., Pachuski, J. et al. (2008) Coenzyme Q 10 supplementation rescues renal disease in Pdss2 kd/kd
mice with mutations in prenyl diphosphate synthase subunit 2. Am. J. Physiol. Physiol. 295, F1535–F1544,
https://doi.org/10.1152/ajprenal.90445.2008
140 Herebian, D., Seibt, A., Smits, S.H.J, B ¨
unning, G., Freyer, C., Prokisch, H. et al. (2017) Detection of 6-demethoxyubiquinone in CoQ10 deficiency
disorders: insights into enzyme interactions and identification of potential therapeutics. Mol. Genet. Metab. 121, 216–223,
https://doi.org/10.1016/j.ymgme.2017.05.012
141 Wang, Y., Oxer, D. and Hekimi, S. (2015) Mitochondrial function and lifespan of mice with controlled ubiquinone biosynthesis. Nat. Commun. 6, 6393,
https://doi.org/10.1038/ncomms7393
142 Herebian, D., Seibt, A., Smits, S.H.J, Rodenburg, R.J., Mayatepek, E. and Distelmaier, F. (2017) 4-Hydroxybenzoic acid restores CoQ 10 biosynthesis in
human COQ2 deficiency. Ann. Clin. Transl. Neurol. 4, 902–908, https://doi.org/10.1002/acn3.486
143 Yubero, D., Montero, R., Martin, M.A., Montoya, J., Ribes, A., Grazina, M. et al. (2016) Secondary coenzyme Q10 deficiencies in oxidative
phosphorylation (OXPHOS) and non-OXPHOS disorders. Mitochondrion 30, 51–58, https://doi.org/10.1016/j.mito.2016.06.007
144 Desbats, M.A., Lunardi, G., Doimo, M., Trevisson, E. and Salviati, L. (2015) Genetic bases and clinical manifestations of Coenzyme Q10 (CoQ10)
deficiency. J. Inherit. Metab. Dis. 38, 145–156, https://doi.org/10.1007/s10545-014-9749-9
145 Sacconi, S., Trevisson, E., Salviati, L., Aym´
e, S., Rigal, O., Garcia Redondo, A. et al. (2010) Coenzyme Q10 is frequently reduced in muscle of patients
with mitochondrial myopathy. Neuromuscul. Disord. 20, 44–48, https://doi.org/10.1016/j.nmd.2009.10.014
146 Montero, R., Grazina, M., L ´
opez-Gallardo, E., Montoya, J., Briones, P., Navarro-Sastre, A. et al. (2013) Coenzyme Q10 deficiency in mitochondrial DNA
depletion syndromes. Mitochondrion 13, 337–341, https://doi.org/10.1016/j.mito.2013.04.001
147 Barca, E., Kleiner, G., Tang, G., Ziosi, M., Tadesse, S., Masliah, E. et al. (2016) Decreased coenzyme Q10 levels in multiple system atrophy cerebellum.
J. Neuropathol. Exp. Neurol. 75, 663–672, https://doi.org/10.1093/jnen/nlw037
148 Hern ´
andez-Camacho, J.D., Bernier, M., L´
opez-Lluch, G. and Navas, P. (2018) Coenzyme Q10 supplementation in aging and disease. Front. Physiol. 9,
44, https://doi.org/10.3389/fphys.2018.00044
149 Marcoff, L. and Thompson, P.D. (2007) The role of coenzyme Q10 in statin-associated myopathy. A systematic review. J. Am. Coll. Cardiol. 49,
2231–2237, https://doi.org/10.1016/j.jacc.2007.02.049
150 Uliˇ
cn ´
a, O., Vanˇ
cov ´
a, O., Waczul´
ıkov ´
a, I., Boˇ
zek, P., ˇ
Sikurov ´
a, L., Bada, V. et al. (2012) Liver mitochondrial respiratory function and coenzyme q content
in rats on a hypercholesterolemic diet treated with atorvastatin. Physiol. Res. 61, 185–193
151 Quinzii, C.M. and Hirano, M. (2011) Primary and secondary CoQ(10) deficiencies in humans. Biofactors 37, 361–365,
https://doi.org/10.1002/biof.155
398 c
2018 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society
... Primary CoQ 10 deficits are autosomal recessive conditions characterised by a wide range of clinical symptoms [53], brought on by biallelic mutations in any of the COQ genes, which biochemically result in decreased levels of CoQ 10 in tissues. The mainly affected organs in primary CoQ deficiencies are the central and peripheral nervous system, heart, skeletal muscle, and kidney [53]. ...
... Primary CoQ 10 deficits are autosomal recessive conditions characterised by a wide range of clinical symptoms [53], brought on by biallelic mutations in any of the COQ genes, which biochemically result in decreased levels of CoQ 10 in tissues. The mainly affected organs in primary CoQ deficiencies are the central and peripheral nervous system, heart, skeletal muscle, and kidney [53]. However, the symptoms and severity of CoQ 10 deficiency can vary widely depending on the specific mutation and the affected genes. ...
... This condition is often observed in primary CoQ 10 deficiency patients with extrarenal manifestations such as sensorineural hearing loss or neurologic deficit [92,93]. Pathogenic variants of PDSS1, PDSS2, COQ2, COQ6, and COQ8B genes have been related to SRNS, alone or in combination with other symptoms [53]. Early use of CoQ 10 supplementation has been shown to minimise proteinuria and slow the disease progression in monogenic SRNS associated with primary CoQ 10 deficiency [94]. ...
Article
Full-text available
Originally identified as a key component of the mitochondrial respiratory chain, Coenzyme Q (CoQ or CoQ10 for human tissues) has recently been revealed to be essential for many different redox processes, not only in the mitochondria, but elsewhere within other cellular membrane types. Cells rely on endogenous CoQ biosynthesis, and defects in this still-not-completely understood pathway result in primary CoQ deficiencies, a group of conditions biochemically characterised by decreased tissue CoQ levels, which in turn are linked to functional defects. Secondary CoQ deficiencies may result from a wide variety of cellular dysfunctions not directly linked to primary synthesis. In this article, we review the current knowledge on CoQ biosynthesis, the defects leading to diminished CoQ10 levels in human tissues and their associated clinical manifestations.
... A central role of CoQ is to act as an electron transporter in mitochondrial respiratory chain (MRC) function [13]. CoQ deficiency is a rare mitochondrial disorder associated with heterogeneous phenotypes and a characteristic decrease in CoQ10 levels in human tissues and plasma [14]. In general, CoQ deficiency due to variants in genes encoding proteins of the CoQ biosynthesis pathway or its regulation is defined as primary CoQ deficiency. ...
... Combined with the results from the burden tests and the fact that all of these variants were ultra-rare (MAF < 0.1% in gnomAD), it is highly likely that a substantial proportion of them are also clinically relevant for different CoQ deficiency phenotypes. However, since the clinical symptoms of mild CoQ deficiency are very often non-specific and CoQ is not chemically stable [14], it can be difficult to determine whether the variant carriers were suffering from CoQ deficiency by content assay of human CoQ10 in forensic samples. ...
Article
Full-text available
Cardiac arrhythmia is currently considered to be the direct cause of death in a majority of sudden unexplained death (SUD) cases, yet the genetic predisposition and corresponding endophenotypes contributing to SUD remain incompletely understood. In this study, we aimed to investigate the involvement of Coenzyme Q (CoQ) deficiency in SUD. First, we re-analyzed the exome sequencing data of 45 SUD and 151 sudden infant death syndrome (SIDS) cases from our previous studies, focusing on previously overlooked genetic variants in 44 human CoQ deficiency-related genes. A considerable proportion of the SUD (38%) and SIDS (37%) cases were found to harbor rare variants with likely functional effects. Subsequent burden testing, including all rare exonic and untranslated region variants identified in our case cohorts, further confirmed the existence of significant genetic burden. Based on the genetic findings, the influence of CoQ deficiency on electrophysiological and morphological properties was further examined in a mouse model. A significantly prolonged PR interval and an increased occurrence of atrioventricular block were observed in the 4-nitrobenzoate induced CoQ deficiency mouse group, suggesting that CoQ deficiency may predispose individuals to sudden death through an increased risk of cardiac arrhythmia. Overall, our findings suggest that CoQ deficiency-related genes should also be considered in the molecular autopsy of SUD.
... 3 The age at onset can vary from infancy to late adulthood. 4 Early supplementation of CoQ10 may result in favorable outcomes in a subset of population, thus a timely diagnosis is essential. 5 We report an interesting case of a young male who presented with slowly progressive cerebellar ataxia and dystonia caused by primary CoQ10 deficiency-4. ...
... Variants in COQ8A represent the most common cause of primary CoQ10 deficiency, also known as autosomal-recessive cerebellar ataxia type-2 (ARCA2) or autosomal recessive spinocerebellar ataxia-9 (SCAR9). 4 The clinical syndrome is characterized by a slowly progressive or an apparently stable ataxia. Central and peripheral nervous system manifestations like epilepsy, cognitive deficits, exercise intolerance, myopathy, are also variably associated with COQ10D4. 5 The movement disorders like myoclonus, dystonia head tremors, postural or action tremors of the limbs, chorea and parkinsonism may complicate the disease course. ...
... 2 Cerebellar ataxia, variably associated with other neurologic and systemic symptoms, is one of the most common clinical presentations of CoQ 10 deficiency. 1 It was initially described in 6 patients in 2001. 3 In 2003, a study 4 reported that approximately 10% of patients with cerebellar ataxia of undefined etiology displayed low levels of CoQ 10 in muscle. ...
... CoQ 10 deficiency in cerebellar ataxia can be primary as well as secondary to mutations in genes unrelated to its biosynthesis. 1,5 The most frequent cause of primary CoQ 10 -deficient cerebellar ataxia is pathogenic variants in COQ8A gene (previously known as ADCK3), 6 while secondary CoQ 10 deficiency has been reported in association with mutations in ANO10, 7 APTX, [8][9][10] SLC25A26, 11 GLUT1, 12 and in several other OxPhos and non-OxPhos disorders. 12 Remarkably, CoQ 10 supplementation is partially effective in both primary and secondary cases. ...
Article
Full-text available
Background and Objectives Coenzyme Q 10 (CoQ 10 )–deficient cerebellar ataxia can be due to pathogenic variants in genes encoding for CoQ 10 biosynthetic proteins or associated with defects in protein unrelated to its biosynthesis. Diagnosis is crucial because patients may respond favorably to CoQ 10 supplementation. The aim of this study was to identify through whole-exome sequencing (WES) the pathogenic variants, and assess CoQ 10 levels, in fibroblasts from patients with undiagnosed cerebellar ataxia referred to investigate CoQ 10 deficiency. Methods WES was performed on genomic DNA extracted from 16 patients. Sequencing data were filtered using a virtual panel of genes associated with CoQ 10 deficiency and/or cerebellar ataxia. CoQ 10 levels were measured by high-performance liquid chromatography in 14 patient-derived fibroblasts. Results A definite genetic etiology was identified in 8 samples of 16 (diagnostic yield = 50%). The identified genetic causes were pathogenic variants of the genes COQ8A ( ADCK3 ) (n = 3 samples), ATP1A3 (n = 2), PLA2G6 (n = 1), SPG7 (n = 1), and MFSD8 (n = 1). Five novel mutations were found ( COQ8A n = 3, PLA2G6 n = 1, and MFSD8 n = 1). CoQ 10 levels were significantly decreased in 3/14 fibroblast samples (21.4%), 1 carrying compound heterozygous COQ8A pathogenic variants, 1 harboring a homozygous pathogenic SPG7 variant, and 1 with an unknown molecular defect. Discussion This work confirms the importance of COQ8A gene mutations as a frequent genetic cause of cerebellar ataxia and CoQ 10 deficiency and suggests SPG7 mutations as a novel cause of secondary CoQ 10 deficiency.
... 177,178 Reportedly, some patients also developed retinopathy or optic atrophy, hypertrophic cardiomyopathy, and sensorineural hearing loss as a result of CoQ10 deficiency. 183 In addition, CoQ10 deficiency is connected with mtDNA point mutations, depletion, and deletions. 184 Nonetheless, Coenzyme Q supplementation was demonstrated to protect against agerelated DNA double-strand breaks and prolong longevity in mice who consumed a polyunsaturated fatty acid (PUFA)-rich diet by attenuating oxidative alterations. ...
Article
Full-text available
Mitochondria are ATP-producing organelles in eukaryotic organisms that serve as the cell’s power plants. Besides, mitochondria are integral to regulating cellular homeostasis and metabolism as a result of their essential roles in reactive oxygen species (ROS) production, bioenergetics, catabolism and anabolism, heme and iron-sulfur biosynthesis, iron and calcium homeostasis, apoptosis and signal transduction, as well as immunity and inflammation. It is well accepted that mitochondria are evolutionarily derived from endosymbiotic alphaproteobacteria within eukaryotic cells adapted for effective energy transduction. Although most of the mitochondrial DNA (mtDNA) is thought to have been transported to the eukaryotic nucleus during evolution, mitochondria may have preserved protein-coding genes within their own DNA. Accumulating data show that a progressive decline of mitochondria regulates aging. The present review aims to outline the role of mitochondria in various aspects of aging, including unfolded protein response, generation of ROS, and the contribution of somatic mtDNA mutations as well as inflammation in aging. Moreover, we propose mitochondria-targeted nanoparticles and mitochondrial genome editing as novel tools to modify mitochondrial genome aberrations.
... CoQ also governs a whole host of different reactions and is tied to many metabolic pathways including fatty acid oxidation, mitochondrial uridine biosynthesis and more recently, of mounting interest, ferroptosis [4][5][6] . Indeed, primary CoQ deficiency is associated to several pathologies including cerebellar ataxia, cardiomyopathy and nephropathy, to name a few 7,8 . ...
Article
Full-text available
Metabolons are protein assemblies that perform a series of reactions in a metabolic pathway. However, the general importance and aptitude of metabolons for enzyme catalysis remain poorly understood. In animals, biosynthesis of coenzyme Q is currently attributed to ten different proteins, with COQ3, COQ4, COQ5, COQ6, COQ7 and COQ9 forming the iconic COQ metabolon. Yet several reaction steps conducted by the metabolon remain enigmatic. To elucidate the prerequisites for animal coenzyme Q biosynthesis, we sought to construct the entire metabolon in vitro. Here we show that this approach, rooted in ancestral sequence reconstruction, reveals the enzymes responsible for the uncharacterized steps and captures the biosynthetic pathway in vitro. We demonstrate that COQ8, a kinase, increases and streamlines coenzyme Q production. Our findings provide crucial insight into how biocatalytic efficiency is regulated and enhanced by these biosynthetic engines in the context of the cell.
... Among these are the production of heme A and coenzyme Q10 (CoQ10, also known as ubiquinone), products of the inhibited mevalonate pathway that are required for mitochondrial electron transport which drives ATP synthesis. CoQ10 deficiencies (whether primary or secondary/acquired) may affect multiple organ systems (including heart and skeletal muscle, as well as other organs that can be affected by COVID-19-lungs, central and peripheral nervous systems, liver, and kidney) [25,26]. Inhibiting the mevalonate pathway, statins lead to dose-dependent reduction in CoQ10. ...
Article
Full-text available
Statins have been widely advocated for use in COVID-19 based on large favorable observational associations buttressed by theoretical expected benefits. However, past favorable associations of statins to pre-COVID-19 infection outcomes (also buttressed by theoretical benefits) were unsupported in meta-analysis of RCTs, RR = 1.00. Initial RCTs in COVID-19 appear to follow this trajectory. Healthy-user/tolerator effects and indication bias may explain these disparities. Moreover, cholesterol drops in proportion to infection severity, so less severely affected individuals may be selected for statin use, contributing to apparent favorable statin associations to outcomes. Cholesterol transports fat-soluble antioxidants and immune-protective vitamins. Statins impair mitochondrial function in those most reliant on coenzyme Q10 (a mevalonate pathway product also transported on cholesterol)—i.e., those with existing mitochondrial compromise, whom data suggest bear increased risks from both COVID-19 and from statins. Thus, statin risks of adverse outcomes are amplified in those patients at risk of poor COVID-19 outcomes—i.e., those in whom adjunctive statin therapy may most likely be given. High reported rates of rhabdomyolysis in hospitalized COVID-19 patients underscore the notion that statin-related risks as well as benefits must be considered. Advocacy for statins in COVID-19 should be suspended pending clear evidence of RCT benefits, with careful attention to risk modifiers.
Preprint
Full-text available
Metabolons are protein assemblies that perform a series of reactions in a metabolic pathway. However, the general importance and aptitude of metabolons for enzyme catalysis remains poorly understood. In animals, biosynthesis of coenzyme Q is currently attributed to ten different proteins with COQ3, COQ4, COQ5, COQ6, COQ7, and COQ9 forming the iconic COQ metabolon. Yet several reaction steps conducted by the metabolon remain enigmatic. To elucidate the prerequisites for animal coenzyme Q biosynthesis, we sought out to construct the entire metabolon in vitro. Here we show that this approach, rooted by ancestral sequence reconstruction, reveals the enzymes responsible for the uncharacterized steps and captures the biosynthetic pathway in vitro . We demonstrate that COQ8, a kinase, increases and streamlines coenzyme Q production. Our findings provide crucial insight into how biocatalytic efficiency is regulated and enhanced by these biosynthetic engines in the context of the cell.
Chapter
Mitochondrial diseases are extremely heterogeneous genetic disorders due to faulty oxidative phosphorylation (OxPhos). No cure is currently available for these conditions, beside supportive interventions aimed at relieving complications. Mitochondria are under a double genetic control carried out by the mitochondrial DNA (mtDNA) and by nuclear DNA. Thus, not surprisingly, mutations in either genome can cause mitochondrial disease. Although mitochondria are usually associated with respiration and ATP synthesis, they play fundamental roles in a large number of other biochemical, signaling, and execution pathways, each being a potential target for therapeutic interventions. These can be classified as general therapies, i.e., potentially applicable to a number of different mitochondrial conditions, or therapies tailored to a single disease, i.e., personalized approaches, such as gene therapy, cell therapy, and organ replacement. Mitochondrial medicine is a particularly lively research field, and the last few years witnessed a steady increase in the number of clinical applications. This chapter will present the most recent therapeutic attempts emerged from preclinical work and an update of the currently ongoing clinical applications. We think that we are starting a new era in which the etiologic treatment of these conditions is becoming a realistic option.
Article
Full-text available
Coenzyme Q10 (CoQ10), a lipophilic substituted benzoquinone, is present in animal and plant cells. It is endogenously synthetized in every cell and involved in a variety of cellular processes. CoQ10 is an obligatory component of the respiratory chain in inner mitochondrial membrane. In addition, the presence of CoQ10 in all cellular membranes and in blood. It is the only endogenous lipid antioxidant. Moreover, it is an essential factor for uncoupling protein and controls the permeability transition pore in mitochondria. It also participates in extramitochondrial electron transport and controls membrane physicochemical properties. CoQ10 effects on gene expression might affect the overall metabolism. Primary changes in the energetic and antioxidant functions can explain its remedial effects. CoQ10 supplementation is safe and well-tolerated, even at high doses. CoQ10 does not cause any serious adverse effects in humans or experimental animals. New preparations of CoQ10 that are less hydrophobic and structural derivatives, like idebenone and MitoQ, are being developed to increase absorption and tissue distribution. The review aims to summarize clinical and experimental effects of CoQ10 supplementations in some neurological diseases such as migraine, Parkinson´s disease, Huntington´s disease, Alzheimer´s disease, amyotrophic lateral sclerosis, Friedreich´s ataxia or multiple sclerosis. Cardiovascular hypertension was included because of its central mechanisms controlling blood pressure in the brainstem rostral ventrolateral medulla and hypothalamic paraventricular nucleus. In conclusion, it seems reasonable to recommend CoQ10 as adjunct to conventional therapy in some cases. However, sometimes CoQ10 supplementations are more efficient in animal models of diseases than in human patients (e.g. Parkinson´s disease) or rather vague (e.g. Friedreich´s ataxia or amyotrophic lateral sclerosis).
Article
Full-text available
Coenzyme Q (ubiquinone or CoQ) is an essential lipid that plays a role in mitochondrial respiratory electron transport and serves as an important antioxidant. In human and yeast cells, CoQ synthesis derives from aromatic ring precursors and the isoprene biosynthetic pathway. Saccharomyces cerevisiae coq mutants provide a powerful model for our understanding of CoQ biosynthesis. This review focusses on the biosynthesis of CoQ in yeast and the relevance of this model to CoQ biosynthesis in human cells. The COQ1-COQ11 yeast genes are required for efficient biosynthesis of yeast CoQ. Expression of human homologs of yeast COQ1-COQ10 genes restore CoQ biosynthesis in the corresponding yeast coq mutants, indicating profound functional conservation. Thus, yeast provides a simple yet effective model to investigate and define the function and possible pathology of human COQ (yeast or human gene involved in CoQ biosynthesis) gene polymorphisms and mutations. Biosynthesis of CoQ in yeast and human cells depends on high molecular mass multisubunit complexes consisting of several of the COQ gene products, as well as CoQ itself and CoQ intermediates. The CoQ synthome in yeast or Complex Q in human cells, is essential for de novo biosynthesis of CoQ. Although some human CoQ deficiencies respond to dietary supplementation with CoQ, in general the uptake and assimilation of this very hydrophobic lipid is inefficient. Simple natural products may serve as alternate ring precursors in CoQ biosynthesis in both yeast and human cells, and these compounds may act to enhance biosynthesis of CoQ or may bypass certain deficient steps in the CoQ biosynthetic pathway.
Article
Full-text available
Insulin resistance in muscle, adipocytes and liver is a gateway to a number of metabolic diseases. Here, we show a selective deficiency in mitochondrial coenzyme Q (CoQ) in insulin-resistant adipose and muscle tissue. This defect was observed in a range of in vitro insulin resistance models and adipose tissue from insulin-resistant humans and was concomitant with lower expression of mevalonate/CoQ biosynthesis pathway proteins in most models. Pharmacologic or genetic manipulations that decreased mitochondrial CoQ triggered mitochondrial oxidants and insulin resistance while CoQ supplementation in either insulin-resistant cell models or mice restored normal insulin sensitivity. Specifically, lowering of mitochondrial CoQ caused insulin resistance in adipocytes as a result of increased superoxide/hydrogen peroxide production via complex II. These data suggest that mitochondrial CoQ is a proximal driver of mitochondrial oxidants and insulin resistance, and that mechanisms that restore mitochondrial CoQ may be effective therapeutic targets for treating insulin resistance.
Article
Full-text available
Coenzyme Q (CoQ) is an essential component of the mitochondrial electron transport chain and an antioxidant in plasma membranes and lipoproteins. It is endogenously produced in all cells by a highly regulated pathway that involves a mitochondrial multiprotein complex. Defects in either the structural and/or regulatory components of CoQ complex or in non-CoQ biosynthetic mitochondrial proteins can result in a decrease in CoQ concentration and/or an increase in oxidative stress. Besides CoQ10 deficiency syndrome and aging, there are chronic diseases in which lower levels of CoQ10 are detected in tissues and organs providing the hypothesis that CoQ10 supplementation could alleviate aging symptoms and/or retard the onset of these diseases. Here, we review the current knowledge of CoQ10 biosynthesis and primary CoQ10 deficiency syndrome, and have collected published results from clinical trials based on CoQ10 supplementation. There is evidence that supplementation positively affects mitochondrial deficiency syndrome and the symptoms of aging based mainly on improvements in bioenergetics. Cardiovascular disease and inflammation are alleviated by the antioxidant effect of CoQ10. There is a need for further studies and clinical trials involving a greater number of participants undergoing longer treatments in order to assess the benefits of CoQ10 treatment in metabolic syndrome and diabetes, neurodegenerative disorders, kidney diseases, and human fertility.
Article
Full-text available
Primary ubiquinone (UQ) deficiency is an important subset of mitochondrial disease that is caused by mutations in UQ biosynthesis genes. To guide therapeutic efforts we sought to estimate the number of individuals who are born with pathogenic variants likely to cause this disorder. We used the NCBI ClinVar database and literature reviews to identify pathogenic genetic variants that have been shown to cause primary UQ deficiency, and used the gnomAD database of full genome or exome sequences to estimate the frequency of both homozygous and compound heterozygotes within seven genetically-defined populations. We used known population sizes to estimate the number of afflicted individuals in these populations and in the mixed population of the USA. We then performed the same analysis on predicted pathogenic loss-of-function and missense variants that we identified in gnomAD. When including only known pathogenic variants, our analysis predicts 1,665 affected individuals worldwide and 192 in the USA. Adding predicted pathogenic variants, our estimate grows to 123,789 worldwide and 1,462 in the USA. This analysis predicts that there are many undiagnosed cases of primary UQ deficiency, and that a large proportion of these will be in developing regions of the world.
Chapter
Vitamin E reacts with lipid peroxyl radical (LOO•) and reduces it to hydroperoxide (LOOH). Vitamin E itself is oxidized and produces vitamin E radical (Reaction 1). (3.1) On the other hand, hydrophilic vitamin C [ascorbate monoanion, (AsH)] is, by itself, a poor antioxidant, but it enhances the antioxidant activity of tocopherols by regenerating the tocopheroxyl to tocopherol (Reaction 2).
Article
Background: Nephrotic syndrome can be caused by a subgroup of mitochondrial diseases classified as primary coenzyme Q10 (CoQ10) deficiency. Pathogenic COQ2 variants are a cause of primary CoQ10 deficiency and present with phenotypes ranging from isolated nephrotic syndrome to fatal multisystem disease. Case-diagnosis/treatment: We report three pediatric patients with COQ2 variants presenting with nephrotic syndrome. Two of these patients had normal leukocyte CoQ10 levels prior to treatment. Pathologic findings varied from mesangial sclerosis to focal segmental glomerulosclerosis, with all patients having abnormal appearing mitochondria on kidney biopsy. In two of the three patients treated with CoQ10 supplementation, the nephrotic syndrome resolved; and at follow-up, both have normal renal function and stable proteinuria. Conclusions: COQ2 nephropathy should be suspected in patients presenting with nephrotic syndrome, although less common than disease due to mutations in NPHS1, NPHS2, and WT1. The index of suspicion should remain high, and we suggest that providers consider genetic evaluation even in patients with normal leukocyte CoQ10 levels, as levels may be within normal range even with significant clinical disease. Early molecular diagnosis and specific treatment are essential in the management of this severe yet treatable condition.
Article
Primary CoQ10 deficiency is a clinically and genetically heterogeneous, autosomal recessive disorder resulting from mutations in genes involved in the synthesis of coenzyme Q10 (CoQ10). To date, mutations in nine proteins required for the biosynthesis of CoQ10 cause CoQ10 deficiency with varying clinical presentations. In 2009 the first patient with mutations in COQ9 was reported in an infant with a neonatal-onset, primary CoQ10 deficiency with multi-system disease. Here we describe four siblings with a previously undiagnosed lethal disorder characterized by oligohydramnios and intrauterine growth restriction, variable cardiomyopathy, anemia, and renal anomalies. The first and third pregnancy resulted in live born babies with abnormal tone who developed severe, treatment unresponsive lactic acidosis after birth and died hours later. Autopsy on one of the siblings demonstrated brain changes suggestive of the subacute necrotizing encephalopathy of Leigh disease. Whole-exome sequencing (WES) revealed the siblings shared compound heterozygous mutations in the COQ9 gene with both variants predicted to affect splicing. RT-PCR on RNA from patient fibroblasts revealed that the c.521 + 2 T > C variant resulted in splicing out of exons 4–5 and the c.711 + 3G > C variant spliced out exon 6, resulting in undetectable levels of COQ9 protein in patient fibroblasts. The biochemical profile of patient fibroblasts demonstrated a drastic reduction in CoQ10 levels. An additional peak on the chromatogram may represent accumulation of demethoxy coenzyme Q (DMQ), which was shown previously to accumulate as a result of a defect in COQ9. This family expands our understanding of this rare metabolic disease and highlights the prenatal onset, clinical variability, severity, and biochemical profile associated with COQ9-related CoQ10 deficiencies.
Article
Background: Coenzyme Q10(CoQ10) is an important component of the mitochondrial respiratory chain (RC) and is critical for energy production. Although the prevalence of CoQ10deficiency is still unknown, the general consensus is that the condition is under-diagnosed. The aim of this study was to retrospectively investigate CoQ10deficiency in frozen muscle specimens in a cohort of ethnically diverse patients who received muscle biopsies for the investigation of a possible RC deficiency (RCD). Methods: Muscle samples were homogenized whereby 600 ×g supernatants were used to analyze RC enzyme activities, followed by quantification of CoQ10by stable isotope dilution liquid chromatography tandem mass spectrometry. The experimental group consisted of 156 patients of which 76 had enzymatically confirmed RCDs. To further assist in the diagnosis of CoQ10deficiency in this cohort, we included sequencing of 18 selected nuclear genes involved with CoQ10biogenesis in 26 patients with low CoQ10concentration in muscle samples. Results: Central 95% reference intervals (RI) were established for CoQ10normalized to citrate synthase (CS) or protein. Nine patients were considered CoQ10deficient when expressed against CS, while 12 were considered deficient when expressed against protein. In two of these patients the molecular genetic cause could be confirmed, of which one would not have been identified as CoQ10deficient if expressed only against protein content. Conclusion: In this retrospective study, we report a central 95% reference interval for 600 ×g muscle supernatants prepared from frozen samples. The study reiterates the importance of including CoQ10quantification as part of a diagnostic approach to study mitochondrial disease as it may complement respiratory chain enzyme assays with the possible identification of patients that may benefit from CoQ10supplementation. However, the anomaly that only a few patients were identified as CoQ10deficient against both markers (CS and protein), while the majority of patients where only CoQ10deficient against one of the markers (and not the other), remains problematic. We therefore conclude from our data that, to prevent possibly not diagnosing a potential CoQ10deficiency, the expression of CoQ10levels in muscle on both CS as well as protein content should be considered.
Article
Coenzyme Q (CoQ) is a redox-active lipid required for mitochondrial oxidative phosphorylation (OxPhos). How CoQ biosynthesis is coordinated with the biogenesis of OxPhos protein complexes is unclear. Here, we show that the Saccharomyces cerevisiae RNA-binding protein (RBP) Puf3p regulates CoQ biosynthesis. To establish the mechanism for this regulation, we employed a multi-omic strategy to identify mRNAs that not only bind Puf3p but also are regulated by Puf3p in vivo. The CoQ biosynthesis enzyme Coq5p is a critical Puf3p target: Puf3p regulates the abundance of Coq5p and prevents its detrimental hyperaccumulation, thereby enabling efficient CoQ production. More broadly, Puf3p represses a specific set of proteins involved in mitochondrial protein import, translation, and OxPhos complex assembly (pathways essential to prime mitochondrial biogenesis). Our data reveal a mechanism for post-transcriptionally coordinating CoQ production with OxPhos biogenesis, and they demonstrate the power of multi-omics for defining genuine targets of RBPs.
Article
Mitoproteases are becoming recognized as key regulators of diverse mitochondrial functions, although their direct substrates are often difficult to discern. Through multi-omic profiling of diverse Saccharomyces cerevisiae mitoprotease deletion strains, we predicted numerous associations between mitoproteases and distinct mitochondrial processes. These include a strong association between the mitochondrial matrix octapeptidase Oct1p and coenzyme Q (CoQ) biosynthesis-a pathway essential for mitochondrial respiration. Through Edman sequencing and in vitro and in vivo biochemistry, we demonstrated that Oct1p directly processes the N terminus of the CoQ-related methyltransferase, Coq5p, which markedly improves its stability. A single mutation to the Oct1p recognition motif in Coq5p disrupted its processing in vivo, leading to CoQ deficiency and respiratory incompetence. This work defines the Oct1p processing of Coq5p as an essential post-translational event for proper CoQ production. Additionally, our data visualization tool enables efficient exploration of mitoprotease profiles that can serve as the basis for future mechanistic investigations.