3670 PART 16 Genes, the Environment, and Disease
causing Kearns-Sayre syndrome (KSS) and the discovery of a point
mutation in ND4, an mtDNA-encoded complex I gene, causing Leber’s
hereditary optic neuropathy (LHON). Following these two discoveries,
>400 pathogenic mtDNA mutations or deletions have been reported to
cause human disease.
MITOCHONDRIAL DNA STRUCTURE AND
FUNCTION
As a result of its circular structure and extranuclear location, the replication and transcription mechanisms of mtDNA differ from the corresponding mechanisms in the nuclear genome, whose nucleosomal
packaging and structure are more complex. Specifically, mitochondria
have their own transcription system, and the mtDNA itself replicates
independently of cellular replication. Because each cell contains many
copies of mtDNA and because the number of mitochondria can vary
during the lifetime of each cell, mtDNA copy number is not directly
coordinated with the cell cycle. Thus, vast differences in mtDNA
copy number are observed between different cell types and tissues
and during the lifetime of a cell. Another important feature of the
mtDNA replication process is a reduced stringency of proofreading
and replication error correction, leading to a greater degree of sequence
variation compared to the nuclear genome. Some of these sequence
variants are silent polymorphisms that do not have the potential for
a phenotypic or pathogenic effect, whereas others may be considered
pathogenic mutations. There are some mutations that may be considered ecogenetic, as they typically remain silent, meaning they do not
cause disease, unless an external event occurs. One classic example is
seen in a common (1:800) mutation in the mitochondrial 12S rRNA
gene, m.A1555G, which is associated with hearing loss but is rapidly
exacerbated by exposure to normal dosages of an aminoglycoside.
Because mtDNA replication is independent of cellular replication, the
percentage of mutant mtDNA copies tend to increase with age in cells
that are terminally differentiated (nonreplicative) at birth such as neurons and myocytes, which may explain some features of mitochondrial
dysfunction with aging.
With respect to transcription, initiation can occur on both strands
and proceeds through the production of an intronless polycistronic
precursor RNA, which is then processed to produce the 13 individual
mRNA and 24 individual tRNA and rRNA products. The 37 mtDNA
genes comprise fully 93% of the 16,569 nucleotides of the mtDNA
in what is known as the coding region. The control region, which is
contained in the D-loop, consists of ~1.1 kilobases (kb) of noncoding
DNA and is thought to have an important role in replication and transcription initiation.
■ MATERNAL INHERITANCE AND LACK OF
RECOMBINATION
In contrast to homologous pair recombination that takes place in the
nucleus, mtDNA molecules do not undergo recombination, such that
mutational events represent the only source of mtDNA genetic diversification. Moreover, it is only the maternal DNA that is transmitted to
the offspring. The fertilized oocyte degrades mtDNA carried from the
sperm in a complex process involving the ubiquitin proteasome system
and autophagy that takes place on the inner membrane of the oocyte.
Thus, although mothers transmit their mtDNA to both their sons and
daughters, only the daughters can transmit the inherited mtDNA to
future generations. Accordingly, mtDNA sequence variation and associated phenotypic traits and diseases are inherited exclusively along
maternal lines, meaning both sons and daughters have equal chances of
having symptomatic disease, with the only significant exception being
LHON, as described below.
The phenotypic expression, including age of onset and the exact
pattern of organ dysfunction, of a pathogenic mtDNA mutation may
vary greatly, even within families. Because of this complex relationship
between mtDNA mutations and disease expression, sometimes it is difficult to recognize the maternal pattern of inheritance at the clinical or
pedigree level. However, evidence of paternal transmission can almost
certainly exclude an mtDNA genetic origin of phenotypic variation or
disease; conversely, a disease affecting both sexes without evidence of
paternal transmission strongly suggests a heritable mtDNA disorder
(Fig. 468-2).
■ MULTIPLE COPY NUMBER (POLYPLOIDY), HIGH
MUTATION RATE, HETEROPLASMY, AND MITOTIC
SEGREGATION
Each aerobic cell in the body has multiple mitochondria, often numbering many hundreds or more in cells with extensive energy production requirements. Furthermore, the number of copies of mtDNA
within each mitochondrion varies from several to hundreds; this is
true of both somatic as well as germ cells, including oocytes in females.
In the case of somatic cells, this means that the impact of most newly
acquired somatic mtDNA mutations is likely to be very small in terms
of total cellular or organ system function; however, because of the
manyfold higher mutation rate during mtDNA replication, numerous
different mutations may accumulate with aging of the organism. It has
been proposed that the total cumulative burden of acquired somatic
mtDNA mutations with age may result in an overall perturbation of
mitochondrial function, contributing to age-related reduction in the
efficiency of oxidative phosphorylation and increased production of
damaging ROS. Because mtDNA (and nDNA) mutations may result
in electron leak within the ETC, the ROS damage may rise above the
normal baseline in some with specific mutations, resulting in increased
susceptibility to somatic mtDNA damage and disease expression. The
accumulation of such acquired somatic mtDNA mutations with aging
may contribute to age-related diseases, such as metabolic syndrome
and diabetes, cancer, and neurodegenerative and cardiovascular disease in any given individual. However, somatic mutations are not
carried forward to the next generation, and the hereditary impact of
mtDNA mutagenesis requires separate consideration of events in the
female germline.
The multiple mtDNA copy number within each cell, including the
maternal germ cells, results in the phenomenon of heteroplasmy, in
contrast to much greater uniformity (homoplasy) of somatic nuclear
DNA sequence. Heteroplasmy for a given mtDNA sequence variant
or mutation arises as a result of the coexistence within a cell, tissue, or
individual of mtDNA molecules bearing more than one version of the
sequence variant (Fig. 468-3). The importance of the heteroplasmy
phenomena to the understanding of mtDNA-related mitochondrial
diseases is critical. The coexistence of mutant and nonmutant (wildtype) mtDNA and the variation of the mutant load, which can be
thought of as the percentage of mutant mtDNA molecules within a
specific cell, tissue, organ, or organism, contribute to the expression of
a phenotype among individuals from the same maternal sibship. At the
level of the oocyte, the percentage of mtDNA molecules bearing each
version of the polymorphic sequence variant or mutation depends on
stochastic events related to partitioning of mtDNA molecules during
the process of oogenesis itself. Thus, oocytes differ from each other
in the degree of heteroplasmy for that sequence variant or mutation.
In turn, the heteroplasmic state is carried forward to the zygote and
to the organism as a whole, to varying degrees, depending on mitotic
segregation of mtDNA molecules during organ system development
and maintenance. For this reason, in vitro fertilization, followed by
preimplantation genetic diagnosis (PGD), is not as predictive of the
genetic health of the offspring in the case of mtDNA mutations as in
the case of mutations and subsequent diseases occurring in the nuclear
genome. Similarly, the impact of somatic mtDNA mutations acquired
during development and subsequently also shows an enormous spectrum of variability. In general, a higher mutant load will result in a
more severe and earlier phenotypic presentation. However, measuring
heteroplasmy in one tissue (lymphocytes from blood or urine sediment
containing kidney and bladder epithelial cells, for example) may not
represent the percentage of mutant heteroplasmy in the tissue or organs
most affected, such as the cardiac atrioventricular node or brain. Furthermore, the threshold of mutant heteroplasmy that results in clinical
illness may vary depending on the specific mutation.
Mitotic segregation refers to the unequal distribution of wild-type
and mutant versions of mtDNA molecules during all cell divisions
that occur during prenatal development and subsequently throughout
Mitochondrial DNA and Heritable Traits and Diseases
3671CHAPTER 468
Oocyte maturation
and mtDNA amplification Fertilization
Mature oocytes
Primary oocytes
Mutant mitochondrion
Normal mitochondrion
Nucleus
Primordial germ
cell containing
mutant mtDNA
High level of mutation
(affected offspring)
Intermediate level
of mutation (mildly
affected offspring)
Low level of mutation
(unaffected offspring)
FIGURE 468-3 Heteroplasmy and the mitochondrial genetic bottleneck. During the production of
primary oocytes, a selected number of mitochondrial DNA (mtDNA) molecules are transferred into
each oocyte. Oocyte maturation is associated with the rapid replication of this mtDNA population.
This restriction-amplification event can lead to a random shift of mtDNA mutational load between
generations and is responsible for the variable levels of mutated mtDNA observed in affected
offspring from mothers with pathogenic mtDNA mutations. Mitochondria that contain mutated
mtDNA are shown in red, and those with normal mtDNA are shown in green. (Reproduced with
permission from R Taylor, D Turnbull: Mitochondrial DNA mutations in human disease. Nat Rev
Genetics 6:389, 2005.)
the lifetime of an individual. The phenotypic effect or disease impact
will be a function not only of the inherent disruptive effect (pathogenicity) on the mtDNA-encoded gene (coding region mutations) or
integrity of the mtDNA molecule (control region mutations) but also
of its distribution among the multiple copies of mtDNA in the various
mitochondria, cells, and tissues of the affected individual. Thus, one
consequence can be the generation of a bottleneck due to the marked
decline in given sets of mtDNA variants, pathogenic and nonpathogenic, consequent to such mitotic segregation. It is postulated that the
main effects of this bottleneck occur between the primordial germ
cell state and the primary oocyte stage of development. Heterogeneity
arises from differences in the degree of heteroplasmy among oocytes of
the transmitting female, together with subsequent, probably random,
mitotic segregation of the pathogenic mutation during tissue and organ
development and throughout the lifetime of the individual offspring.
The actual expression of disease might then depend on a threshold
percentage of mitochondria whose function is disrupted by mtDNA
mutations. This in turn confounds hereditary transmission patterns
and hence genetic diagnosis of pathogenic heteroplasmic mutations.
Generally, if the proportion of mutant mtDNA is <60%, the individual
is unlikely to be affected, whereas proportions exceeding 90% cause
clinical disease. One notable exception is LHON, in which these mutations are present either in 100% mutant homoplasmy, which causes the
disease expression, or 100% wild-type homoplasmy. It is not understood why this specific phenotype and the several known mtDNA
alleles that result in LHON behave in this manner.
■ HOMOPLASMIC VARIANTS AND HUMAN mtDNA
PHYLOGENY
In contrast to classic mtDNA diseases, most of which begin in childhood
and are the result of heteroplasmic mutations as noted above, during
the course of human evolution, certain mtDNA sequence variants have
drifted to a state of homoplasmy, wherein all of the mtDNA molecules
in the organism contain the new sequence variant. This arises due to a
“bottleneck” effect followed by genetic drift during the very process of
oogenesis itself (Fig. 468-3). In other words, during certain stages of
oogenesis, the mtDNA copy number becomes so substantially reduced
that the particular mtDNA species bearing the novel or derived
sequence variant may become the increasingly predominant, and eventually exclusive, version of the mtDNA for
that particular nucleotide site. All of the offspring of a
woman bearing an mtDNA sequence variant or mutation
that has become homoplasmic will also be homoplasmic
for that variant and will transmit the sequence variant
forward in subsequent generations.
Considerations of reproductive fitness limit the evolutionary or population emergence of pathogenic homoplasmic mutations that are lethal or cause severe disease
in infancy or childhood. Thus, with a number of notable
exceptions (e.g., mtDNA mutations causing LHON; see
below), most homoplasmic mutations are considered to
be neutral markers of human evolution, which are useful
and interesting in the population genetics analysis of
shared maternal ancestry but have little significance in
human phenotypic variation or disease predisposition.
More important is the understanding that this accumulation of homoplasmic mutations occurs at a genetic
locus that is transmitted only through the female germline and that lacks recombination. In turn, this enables
reconstruction of the sequential topology and radiating
phylogeny of mutations accumulated through the course
of human evolution since the time of the most recent
common mtDNA ancestor of all contemporary mtDNA
sequences, some 200,000 years ago. The term haplogroup
is usually used to define major branching points in
the human mtDNA phylogeny, nested one within the
other, which often demonstrate striking continental geographic ancestral partitioning. At the level of the complete mtDNA sequence, the term haplotype is usually
used to describe the sum of mutations observed for a given mtDNA
sequence and as compared to a reference sequence, such that all haplotypes falling within a given haplogroup share the total sum of mutations that have accumulated since the most recent common ancestor
and the bifurcation point they mark. The remaining observed variants
are private to each haplotype. Consequentially, the human mtDNA
sequence is an almost perfect molecular prototype for a nonrecombining locus, and its variation has been extensively used in phylogenetic
studies. Moreover, the mtDNA mutation rate is considerably higher
than the rate observed for the nuclear genome, especially in the control region, which contains the displacement loop, or D-loop, in turn
comprising two adjacent hypervariable regions (HVR-I and HVR-II).
Together with the absence of recombination, this amplifies drift to
high frequencies of novel haplotypes. As a result, mtDNA haplotypes
are more highly partitioned across geographically defined populations
than sequence variants in other parts of the genome. Despite extensive
research, it has not been well established that such haplotype-based
partitioning has a significant influence on human health conditions.
However, mtDNA-based phylogenetic analysis can be used both as a
quality assurance tool and as a filter in distinguishing neutral mtDNA
variants comprising human mtDNA phylogeny from potentially deleterious mutations.
MITOCHONDRIAL DNA DISEASE
The true prevalence of mtDNA disease is difficult to estimate because
of the phenotypic heterogeneity that occurs as a function of heteroplasmy, the challenge of detecting and assessing heteroplasmy in
different affected tissues, and the other unique features of mtDNA
function and inheritance described above. It is estimated that at least
1 in 200 healthy humans harbors a pathogenic mtDNA mutation with
the potential to causes disease but that heteroplasmic germline pathogenic mtDNA mutations actually result in clinical disease in ~1 in 5000
individuals.
The true disease burden relating to mtDNA sequence variation will
only be known when the following capabilities become available: (1)
ability to distinguish a completely neutral sequence variant from a true
phenotype-modifying or pathogenic mutation, (2) accurate assessment of heteroplasmy that can be determined with fidelity, and (3) a
3672 PART 16 Genes, the Environment, and Disease
Parkinsonism,
aminoglycoside-induced deafness
MELAS
myoglobinuria Myopathy,
PEO
Cardiomyopathy
ECM
Diabetes and deafness
Cardiomyopathy,
SIDS, ECM Cardiomyopathy,
PEO, MERRF,
MELAS, deafness
PPK, deafness,
MERRF-MELAS
PEO
PEO
Myopathy, lymphoma
Myopathy, MELAS
Myopathy,
cardiomyopathy, PEO
PEO, LHON, MELAS,
myopathy, cardiomyopathy,
diabetes and deafness
Cardiomyopathy
LS, MELAS,
multisystem disease
ECM
Myopathy,
PEO
LS, ataxia,
chorea, myopathy
Cardiomyopathy
myoclonus
Cardiomyopathy
ECM
Cardiomyopathy, ECM
PEO, myopathy,
sideroblastic anemia
ECM, LHON, myopathy,
cardiomyopathy, MELAS
and parkinsonism
LHON, MELAS,
diabetes,
LHON and dystonia
LS, MELAS
LHON, myopathy,
LHON and dystonia
LHON
LS, ECM,
myoglobinuria
NARP, MILS,
FBSN
Cardiomyopathy
LHON
LHON Cyt b
ND6
ND2 ND5
ND4
ND4L
ND3
COXIII
COXII
COXI
ND1
L1
16S
V 12sF
PT
E
H
R
K G
D
S1
Y
C
N
A
W
M
Q
I
L2
S2
A6
A8
Myoglobinuria,
motor neuron disease,
sideroblastic anemia
Myopathy,
multisystem disease,
encephalomyopathy
Progressive myoclonus,
epilepsy, and optic atrophy
FIGURE 468-4 Mutations in the human mitochondrial genome known to cause disease. Disorders that are frequently
or prominently associated with mutations in a particular gene are shown in boldface. Diseases due to mutations that
impair mitochondrial protein synthesis are shown in blue. Diseases due to mutations in protein-coding genes are shown
in red. ECM, encephalomyopathy; FBSN, familial bilateral striatal necrosis; LHON, Leber’s hereditary optic neuropathy;
LS, Leigh’s syndrome; MELAS, mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes; MERRF,
myoclonic epilepsy with ragged red fibers; MILS, maternally inherited Leigh’s syndrome; NARP, neuropathy, ataxia,
and retinitis pigmentosa; PEO, progressive external ophthalmoplegia; PPK, palmoplantar keratoderma; SIDS, sudden
infant death syndrome. (From S DiMauro, E Schon: Mitochondrial respiratory-chain diseases. N Engl J Med 348:2656,
2003. Copyright © 2003, Massachusetts Medical Society. Reprinted with permission from Massachusetts Medical
Society.)
systems biology approach (Chap. 486) to
determine the network of epistatic interactions of mtDNA sequence variations
with mutations in the nuclear genome.
■ OVERVIEW OF CLINICAL
AND PATHOLOGIC FEATURES
OF HUMAN mtDNA DISEASE
Given the vital roles of mitochondria
in all nucleated cells, it is not surprising that mtDNA mutations can affect
numerous tissues with pleiotropic
effects. More than 200 different diseasecausing, mostly heteroplasmic mtDNA
mutations have been described affecting
ETC function. Figure 468-4 provides
a partial mtDNA map of some of the
better characterized of these disorders.
A number of clinical clues can increase
the index of suspicion for a heteroplasmic mtDNA mutation as an etiology
of a heritable trait or disease, including
(1) familial clustering with absence of
paternal transmission; (2) adherence to
one of the classic syndromes (see below)
or paradigmatic combinations of disease
phenotypes involving several organ systems that normally do not fit together
within a single nuclear genomic mutation category; (3) a complex of laboratory and pathologic abnormalities that
reflect disruption in cellular energetics
(e.g., lactic acidosis and neurodegenerative and myodegenerative symptoms
with the finding of ragged red fibers,
reflecting the accumulation of abnormal mitochondria under the muscle sarcolemmal membrane); or (4) a mosaic
pattern reflecting a heteroplasmic state.
There are no truly sensitive and specific biomarkers of disease, and the
presence of a historically quintessential finding of ragged red fibers can
be seen in numerous muscle disorders, so laboratory tests must always
be interpreted in the context of their limitations and should not be used
to define the disease.
Heteroplasmy can sometimes be elegantly demonstrated at the
tissue level using histochemical staining for enzymes in the oxidative
phosphorylation pathway, with a mosaic pattern indicating heterogeneity of the genotype for the coding region for the mtDNA-encoded
enzyme. Complex II, CoQ, and cytochrome c are exclusively encoded
by nuclear DNA. In contrast, complexes I, III, IV, and V contain at least
some subunits encoded by mtDNA. Just 3 of the 13 subunits of the
ETC complex IV enzyme, cytochrome c oxidase (COX), are encoded
by mtDNA, and therefore, this enzyme has the lowest threshold for
dysfunction when a threshold level of mutated mtDNA is reached. Histochemical staining for COX activity in tissues of patients affected with
heteroplasmic inherited mtDNA mutations (or with the somatic accumulation of mtDNA mutations, see below) can show a mosaic pattern
of reduced histochemical staining in comparison with histochemical
staining for the complex II enzyme succinate dehydrogenase (SDH)
(Fig. 468-5). Heteroplasmy can also be detected at the genetic level
through direct Sanger-type mtDNA genotyping under special conditions, although clinically significant low levels of heteroplasmy can
escape detection in genomic samples extracted from whole blood using
conventional genotyping and sequencing techniques. Next-generation
sequencing (NGS) has largely overcome these limitations and enables
reliable detection and quantitation of heteroplasmy.
NGS has dramatically improved the clinical genetic diagnostic evaluation of mitochondrial diseases at the level of both the nuclear genome
and mtDNA. In the context of the larger nuclear genome, the ability
of NGS techniques to dramatically increase the speed at which DNA
can be sequenced at a fraction of the cost of conventional Sanger-type
sequencing technology is particularly beneficial. Low sequencing costs
and short turnaround time expedite “first-tier” screening of panels of
hundreds of previously known or suspected mitochondrial disease genes
or screening for the entire exome or genome in an attempt to identify
novel genes and mutations affecting different patients or families. In the
context of the mtDNA, NGS approaches now provide rapid and reliable
detection of heteroplasmy in different affected tissues. Although Sanger
sequencing allows for complete coverage of the mtDNA, it is limited by
the lack of deep coverage and low sensitivity for heteroplasmy detection
when levels are <50%. In contrast, NGS technology is an excellent tool
for rapidly and accurately obtaining a patient’s predominant mtDNA
sequence as well as lower frequency heteroplasmic variants and can
reliably detect mutant heteroplasmy <10%. Lower levels are often only
clinically relevant if in the setting of a striking difference in heteroplasmy
in different tissues. This capability to detect heteroplasmy at levels that
assist in pointing to an mtDNA-based disease process emanates from
deep coverage of the genome through multiple independent sequence
reads. Accordingly, recent studies making use of NGS techniques have
demonstrated sequence accuracy equivalent to Sanger-type sequencing
but also have uncovered heretofore unappreciated heteroplasmy rates
ranging between 10% and 50% and detection of single nucleotide heteroplasmy down to levels of <10%.
Clinically, the most striking overall characteristic of mitochondrial
genetic disease is the phenotypic heterogeneity associated with mtDNA
mutations. This extends to intrafamilial phenotypic heterogeneity
for the same mtDNA pathogenic mutation and, conversely, to the
overlap of phenotypic disease manifestations with distinct mutations.
Thus, although fairly consistent and well-defined “classic” syndromes
Mitochondrial DNA and Heritable Traits and Diseases
3673CHAPTER 468
FIGURE 468-5 Cytochrome c oxidase (COX) deficiency in mitochondrial DNA (mtDNA)–associated disease. Transverse
tissue sections that have been stained for COX and succinate dehydrogenase (SDH) activities sequentially, with
COX-positive cells shown in brown and COX-deficient cells shown in blue. A. Skeletal muscle from a patient with a
heteroplasmic mitochondrial tRNA point mutation. The section shows a typical “mosaic” pattern of COX activity, with
many muscle fibers harboring levels of mutated mtDNA that are above the crucial threshold to produce a functional
enzyme complex. B. Cardiac tissue (left ventricle) from a patient with a homoplasmic tRNA mutation that causes
hypertrophic cardiomyopathy, which demonstrates an absence of COX in most cells. C. A section of cerebellum from a
patient with mtDNA rearrangement that highlights the presence of COX-deficient neurons. D, E. Tissues that show COX
deficiency due to clonal expansion of somatic mtDNA mutations within single cells—a phenomenon that is seen in both
postmitotic cells (D; extraocular muscles) and rapidly dividing cells (E; colonic crypt) in aging humans. (Reproduced
with permission from R Taylor, D Turnbull: Mitochondrial DNA mutations in human disease. Nat Rev Genetics 6:389,
2005.)
have been attributed to specific mutations, frequently “nonclassical”
combinations of disease phenotypes ranging from isolated myopathy
to extensive multisystem disease are often encountered, rendering
genotype-phenotype correlation challenging. In both classical and
nonclassical mtDNA disorders, there is often a clustering of some combination of abnormalities affecting the neurologic system (including
optic nerve atrophy, pigment retinopathy, and sensorineural hearing
loss), cardiac and skeletal muscle (including extraocular muscles), and
endocrine and metabolic systems (including diabetes mellitus). Additional organ systems that may be affected include the hematopoietic,
renal, hepatic, and gastrointestinal systems, although these are more
frequently involved in infants and children. Disease-causing mtDNA
coding region mutations can affect either one of the 13 proteinencoding genes or one of the 24 protein synthetic genes. Clinical manifestations do not readily distinguish these two categories, although
lactic acidosis and specific muscle pathologic findings (e.g., ragged
red and ragged blue fibers, described immunohistochemical staining,
paracrystalline inclusions on ultrastructure) tend to be more prominent in the latter. In all cases, either defective ATP production due
to disturbances in the ETC or enhanced generation of ROS has been
invoked as the mediating biochemical mechanism between mtDNA
mutation and disease manifestation.
■ mtDNA DISEASE PRESENTATIONS
The clinical presentation of adult patients with mtDNA disease can
be divided into three categories: (1) clinical features suggestive of
mitochondrial disease (Table 468-2) but not a well-defined classic
syndrome; (2) classic mtDNA syndromes; and (3) clinical presentation
confined to one organ system (e.g., isolated sensorineural deafness,
cardiomyopathy, or diabetes mellitus).
Table 468-3 provides a summary of eight illustrative classic mtDNA
syndromes or disorders that affect adult patients and highlights some
of the most interesting features of mtDNA disease in terms of molecular pathogenesis, inheritance, and clinical presentation. The first five
of these syndromes result from heritable point mutations in either
protein-encoding or protein synthetic mtDNA genes; the other three
result from rearrangements or deletions
that usually do not involve the germline.
LHON is a common cause of maternally inherited visual failure. LHON typically presents during young adulthood
with subacute painless loss of vision in
one eye, with symptoms developing in
the other eye 6–12 weeks later. In some
instances, cerebellar ataxia, peripheral
neuropathy, and cardiac conduction
defects are observed. In >95% of cases,
LHON is due to one of the three homoplasmic point mutations of mtDNA that
affect genes encoding different subunits
of complex I of the mitochondrial ETC;
however, not all individuals who inherit
a primary LHON mtDNA mutation
develop optic neuropathy, and the maleto-female ratio is 8.2, indicating that
additional environmental (e.g., tobacco
exposure) or independent genetic factors are important in the etiology of the
disorder. Both the nuclear and mitochondrial genomic backgrounds modify
disease penetrance. Indeed, a region of
the X chromosome containing a highrisk haplotype for LHON has been
identified, supporting the formulation
that nuclear genes act as modifiers and
affording an explanation for the male
prevalence of LHON. This haplotype
can be used in predictive genomic testing
and prenatal screening for this disease.
In contrast to the other classic mtDNA disorders, it is of interest that
patients with this syndrome are often homoplasmic for the diseasecausing mutation. The somewhat later onset in young adulthood and
modifying effect of protective background nuclear genomic haplotypes
may have enabled homoplasmic pathogenic mutations to have escaped
evolutionary censoring.
Mitochondrial encephalomyopathy, lactic acidosis, and stroke-like
episodes (MELAS) is a multisystem disorder with a typical onset
between 2 and 10 years of age. Following normal early psychomotor
development, the most common initial symptoms are seizures, recurrent headaches, anorexia, and recurrent vomiting. Exercise intolerance
or proximal limb weakness can be the initial manifestation, followed
by generalized tonic-clonic seizures. Short stature is common. Seizures
are often associated with stroke-like episodes of transient hemiparesis
or cortical blindness that may produce recurrent encephalopathy with
impaired consciousness. It is often not possible to determine if the
encephalopathy is due to refractory clinical or subclinical seizures or
should be attributed to an independent effect. The cumulative residual
effects of the stroke-like episodes gradually impair motor abilities,
vision, and cognition, often by adolescence or young adulthood.
Sensorineural hearing loss adds to the progressive decline of these
TABLE 468-2 Common Features of Mitochondrial DNA–Associated
Diseases in Adults
Neurologic: stroke, epilepsy, migraine headache, peripheral neuropathy, ataxia,
dystonia, myoclonus, cranial neuropathy (optic atrophy, sensorineural deafness,
dysphagia, dysphasia)
Skeletal myopathy: ophthalmoplegia, exercise intolerance, myalgia, weakness
Cardiac: conduction block, cardiomyopathy
Respiratory: hypoventilation, aspiration pneumonitis
Endocrine: diabetes mellitus, premature ovarian failure, hypothyroidism,
hypoparathyroidism
Ophthalmologic: cataracts, pigment retinopathy, neurologic and myopathic (optic
atrophy, ophthalmoplegia)
3674 PART 16 Genes, the Environment, and Disease
individuals. A plethora of less common symptoms have been described
including myoclonus, ataxia, episodic coma, optic atrophy, cardiomyopathy, pigmentary retinopathy, ophthalmoplegia, diabetes mellitus,
hirsutism, gastrointestinal dysmotility, and nephropathy. The typical
age of death ranges from 10 to 35 years, but some individuals live into
their sixth decade. Intercurrent infections or intestinal obstructions are
often the terminal events. It is not atypical for some family members
to have much less severe or later onset illness, presumably because of
a lessor mutation load, and “MELAS” is not used as a diagnosis for
these restricted phenotypes. This creates somewhat of a disconnect
between the genotype for MELAS (most commonly the m.3243A>G
mutation) and a diverse phenotype, which includes the syndrome
MELAS, a syndrome of high-frequency hearing loss and diabetes with
onset later in life, as well as many other phenotypes between these two
extreme syndromes. Certain other mtDNA mutations can also cause
such patterns of diverse phenotypic expression. Laboratory investigation commonly demonstrates elevated lactate concentrations at rest
with excessive increase after moderate exercise. Brain imaging during
stroke-like episodes shows areas of involvement on T2- or fluid-attenuated inversion recovery (FLAIR) sequences, with decreased signal on
perfusion-weighted sequences, which typically involve the posterior
cerebrum and do not conform to the distribution of major arteries.
These magnetic resonance imaging (MRI) abnormalities may be temporary or evolve to subsequent atrophy (Fig. 468-6). Electrocardiography (ECG) may show evidence of cardiomyopathy, preexcitation, or
incomplete heart block. Electromyography and nerve conduction studies are consistent with a myopathic process, without or with coexisting
axonal and sensory neuropathic findings. Muscle biopsy typically
shows ragged red fibers with the modified Gomori trichrome stain or
“ragged blue fibers” with the SDH histochemical stain, resulting from
the hyperintense reaction. The diagnosis of MELAS is based on a combination of clinical findings and molecular genetic testing. Mutations
in the mtDNA gene MT-TL1 encoding tRNAleu are causative. The most
common mutation, present in ~80% of individuals with typical clinical
findings, is an A-to-G transition at nucleotide 3243 (m.3243A>G).
Mutations can usually be detected in mtDNA from leukocytes in individuals with typical MELAS; however, the occurrence of heteroplasmy
can result in varying tissue distribution of mutated mtDNA. In the
absence of specific treatment, various manifestations of MELAS are
treated according to standard modalities for prevention, surveillance,
and treatment. Recent developments in therapy are described below.
Myoclonus epilepsy with ragged red fiber (MERRF) is a multisystem
disorder characterized by myoclonus, seizures, ataxia, and myopathy
with ragged red fibers. Hearing loss, exercise intolerance, neuropathy,
ataxia, cervical lipomas, and short stature are often present. Ataxia and
lipomas can be a feature in adults or adult-onset MERRF. Cerebrospinal fluid (CSF) analysis reveals an elevated protein content. Almost all
MERRF patients have a mutation in the mtDNA tRNAlys gene, and the
m.8344A>G mutation in the mtDNA gene encoding the lysine amino
acid tRNA is responsible for 80–90% of MERRF cases.
Neuropathy, ataxia, and retinitis pigmentosa (NARP) is characterized by moderate diffuse cerebral and cerebellar atrophy and symmetric lesions of the basal ganglia on MRI (Figs. 468-7 and 468-8). A
heteroplasmic m.8993T>G mutation in the ATPase 6 subunit gene has
been identified as causative, which underscores the lack of definitive
genotype-phenotype correlation in mitochondrial diseases. Ragged
red fibers are not observed in muscle biopsy. When >95% of mtDNA
TABLE 468-3 Mitochondrial Diseases Due to Mitochondrial DNA (mtDNA) Point Mutations and Large-Scale Rearrangements
DISEASE PHENOTYPE
MOST FREQUENT mtDNA
MUTATIONS
HETEROPLASMIC/
HOMOPLASMIC MATERNAL
NARP, Leigh’s syndrome Loss of central vision leading to blindness in young
adult life
m.1778G>A, m.14484T>C,
m.3460G>A
Heteroplasmic Maternal
MELAS Mitochondrial encephalomyopathy, lactic acidosis,
and stroke-like episodes; may manifest only as
diabetes mellitus
Point mutation in tRNAleu Heteroplasmic Maternal
MERRF Myoclonic epilepsy, ragged red fibers in muscle,
ataxia, increased CSF protein, sensorineural deafness,
dementia
Point mutation in tRNAlys Heteroplasmic Maternal
Deafness Progressive sensorineural deafness, often induced by
aminoglycoside antibiotics
m.1555A>G mutation in 12S
rRNA
Homoplasmic Maternal
Nonsyndromic sensorineural deafness m.7445A>G mutation in 12S
rRNA
Homoplasmic Maternal
Chronic progressive
external
ophthalmoplegia (PEO)
Late-onset bilateral ptosis and ophthalmoplegia,
proximal muscle weakness, and exercise intolerance
Single deletions or duplications Heteroplasmic Mostly sporadic, somatic
mutations
Pearson’s syndrome Pancreatic insufficiency, pancytopenia, lactic acidosis Large deletion Heteroplasmic Sporadic, somatic mutations
Kearns-Sayre syndrome
(KSS)
External ophthalmoplegia, heart block, retinal
pigmentation, ataxia
The 5-kb “common deletion” Heteroplasmic Sporadic, somatic mutations
Abbreviations: CSF, cerebrospinal fluid; NARP, neuropathy, ataxia, and retinitis pigmentosa.
FIGURE 468-6 A 15-year-old girl with MELAS (mitochondrial encephalomyopathy,
lactic acidosis, and stroke-like episodes) due to m.A3243G (tRNALeu(UUR)), 85%
mutant heteroplasmy, presenting at age 5 with focal motor seizures, ataxia, and
short stature, with episodes of acute language and motor dysfunction and progress
cognitive impairment. The fluid-attenuated inversion recovery (FLAIR) MRI shows
increased signal intensity (white arrows) in the left temporal-parietal region in
addition to global mild volume loss (increased extra-axial cerebrospinal fluid
spaces).
Mitochondrial DNA and Heritable Traits and Diseases
3675CHAPTER 468
FIGURE 468-7 A 9-year-old girl with Leigh’s syndrome due to m.T8993G (ATPase
subunit 6), 99% heteroplasmy, presenting at age 14 months with a motor delay and
who underwent an MRI at 24 months, at which time she had just begun to walk. She
has moderate cognitive impairment, arm chorea, and distal leg dystonia. The fluidattenuated inversion recover (FLAIR) MRI shows symmetric bilateral increased
signal in the caudate nuclei (thin arrow) and putamen (thick arrow); only left-sided
lesions indicated with arrows.
FIGURE 468-8 A 12-year-old boy with Leigh’s syndrome due to m.T10191C (ND3
gene, complex I), heteroplasmy percentage not determined, presenting with
infantile spasms at 8 months of life. He responded well to adrenocorticotropic
hormone (ACTH), and his MRI and development were normal until 30 months when
he developed dystonia and progressive medically intractable epilepsy. The fluidattenuated inversion recover (FLAIR) MRI at 6 years of life shows global atrophy
with large extra-axial cerebrospinal fluid spaces, increased signal intensity in the
cortex (thin arrows), necrotic bilaterally symmetric lesions in the putamina, and
enlarged lateral ventricles due to loss of bilateral caudate nuclei volume (stars).
molecules are mutant, a more severe clinical, neuroradiologic, and neuropathologic picture (Leigh’s syndrome) emerges. Not uncommonly,
an infant is diagnosed with Leigh’s syndrome due to the m.8993T>G
mutation and not until several years later will the mother present with
symptoms of NARP, a situation that highlights the concept of a higher
threshold for lower levels of tissue heteroplasmy.
Point mutations in the mtDNA gene encoding the 12S rRNA
(m.A1555G) result in heritable nonsyndromic hearing loss. One such
mutation causes heritable ototoxic susceptibility to standard dosing of
aminoglycoside antibiotics, which opens a pathway for a simple pharmacogenetic test in the appropriate clinical settings. This is an example
of an ecogenetic disorder in that most people with this mutation do not
develop any symptoms until exposed to an external agent.
KSS, sporadic progressive external ophthalmoplegia (PEO), and
Pearson’s syndrome are three disease phenotypes caused by large-scale
mtDNA rearrangements including partial deletions or partial duplication. The majority of single large-scale rearrangements of mtDNA are
thought to result from clonal amplification of a single sporadic mutational event, occurring in the maternal oocyte during early embryonic
development. The typical mtDNA deletion specifically involves 4977
nucleotides, accounting for most KSS and PEO of mtDNA deletion
origin. Because germline involvement is rare, most cases are sporadic rather than inherited. KSS is characterized by the triad of onset
before age 20, chronic PEO, and pigmentary retinopathy. Cerebellar
syndrome, heart block, increased CSF protein content, diabetes mellitus, and short stature are also part of the syndrome. Single deletions/
duplication can also result in milder phenotypes such as PEO, characterized by late-onset PEO, proximal myopathy, and exercise intolerance. In
both KSS and PEO, diabetes mellitus and hearing loss are frequent accompaniments. Pearson’s syndrome is also characterized by infantile onset of
a sideroblastic anemia accompanied by lactic acidosis and failure to thrive
caused in part by exocrine pancreatic insufficiency. If the child survives,
the manifestations appear phenotypically similar to those of severe KSS
with myopathy, PEO, encephalopathy, and cardiomyopathy. Pearson’s
syndrome is generally caused by large-scale sporadic deletion of several
mtDNA genes that differ from the common deletion seen in KSS. Typically, the deletion size is larger in Pearson’s syndrome, and located with
different break points, than in KSS or PEO, but this is not always the case.
Two important dilemmas in classic mtDNA disease have benefited
from recent important research insights. The first relates to the greater
involvement of neuronal, muscular, renal, hepatic, and pancreatic manifestations in mtDNA disease in these syndromes. This observation
has appropriately been mostly attributed to the high energy utilization
of the involved tissues and organ systems and, hence, greater dependency on mitochondrial ETC integrity and health. However, because
mutations are stochastic events, mitochondrial mutations should
occur in any organ during embryogenesis and development. Recently,
additional explanations have been suggested based on studies of the
common m.3243A>G transition. The proportion of this mutation in
peripheral blood cells was shown to decrease exponentially with age. A
selective process acting at the stem cell level with a strong bias against
the mutated form would have its greatest effect to reduce the mutant
mtDNA only in highly proliferating cells, such as those derived from
the hematopoietic system. Tissues and organs carrying pathogenic
mtDNA mutations and having a lower cell turnover, such as brain,
nerve, or retina, would not benefit from this effect and, thus, would
accumulate mutational load and be the most affected.
The other dilemma arises from the observation that only a subset of mtDNA mutations accounts for the majority of the familial
mtDNA diseases. The random occurrence of mutations in the mtDNA
sequence should yield a more uniform distribution of disease-causing
mutations. However, recent studies using the introduction of one
severe and one mild point mutation into the female germline of experimental animals demonstrated selective elimination during oogenesis
of the severe mutation and selective retention of the milder mutation,
with the emergence of mitochondrial disease in offspring after multiple
generations. Thus, oogenesis itself can act as an “evolutionary” filter for
the most harmful mtDNA disease.
■ THE INVESTIGATION OF SUSPECTED mtDNA
DISEASE
The clinical presentations of classic syndromes, groupings of disease
manifestations in multiple organ systems, or unexplained isolated
3676 PART 16 Genes, the Environment, and Disease
presentations of one of the disease features of a classic mtDNA syndrome should prompt a systematic clinical investigation as outlined in
Fig. 468-9. Indeed, mitochondrial disease should be considered in the
differential diagnosis of any progressive multisystem disorder. Despite
the centrality of disruptive oxidative phosphorylation, an elevated
blood lactate level is neither specific nor sensitive, because there are
many causes of blood lactic acidosis and many patients with mtDNA
defects presenting at any age may have normal blood lactate levels. An
elevated CSF lactate is a more specific test for mitochondrial disease if
there is central nervous system involvement. The serum creatine kinase
may be elevated but is often normal, even in the presence of a proximal
myopathy. Recently, testing for elevated levels of growth differentiating
factor 15 (GDF15) has shown a high degree of sensitivity and specificity in those with a mitochondrial myopathy, but the degree of elevation
for an individual patient reflects the severity of the illness and does
not seem to be a sensitive marker of disease activity. Urinary organic
acids (specifically TCA cycle intermediates) and amino acids (alanine,
proline) may also be abnormal, reflecting metabolic as well as kidney
proximal tubule dysfunction. Every patient with seizures, episodes of
confusion or atypical behavioral changes, or cognitive decline should
have an electroencephalogram. A brain computed tomography (CT)
scan may show calcified basal ganglia or bilateral hypodense regions
with cortical atrophy. MRI is indicated in patients with brainstem signs
or stroke-like episodes.
For some mitochondrial diseases, it is possible to obtain an accurate
diagnosis with a simple molecular genetic screen. For examples, 95%
of patients with LHON harbor one of the three mtDNA point mutations (m.11778A>G, m.A3460A>G, or m.14484T>C). These patients
have very high levels of mutated mtDNA in peripheral blood cells, and
therefore, it is appropriate to send a blood sample for molecular genetic
analysis by polymerase chain reaction (PCR) or restriction fragment
length polymorphism (RFLP). The same is true for most MERRF
patients who harbor a point mutation in the lysine tRNA gene at position 8344. In contrast, patients with the m.3243A>G MELAS mutation
often have low levels of mutated mtDNA in blood. If clinical suspicion
is strong enough to warrant peripheral blood testing, then patients with
a negative result should have testing repeated using a saliva sample
or be investigated further by performing a skeletal muscle biopsy to
obtain mtDNA from a relatively nonreplicative tissue.
Muscle biopsy histochemical analysis had been the historical cornerstone for investigation of patients with suspected mitochondrial
disease. Histochemical analysis may show subsarcolemmal accumulation of mitochondria with the appearance of ragged red fibers, especially in those with mtDNA mutations affecting the tRNA and rRNA
genes. Electron microscopy might show abnormal mitochondria with
paracrystalline inclusions. Muscle histochemistry may show COXdeficient fibers, which indicate mitochondrial dysfunction (Fig. 468-5).
Respiratory chain complex assays may also show reduced enzyme
function. If enzymatic or polarographic data are used to aid in the
confirmation of diagnosis, a standard method of analysis should be
employed. Either of these two abnormalities, within the exact context
of established peer-reviewed criteria, may confirm the presence of a
mitochondrial disease, to be followed by an in-depth molecular genetic
analysis. In most major centers, genetic testing has become the primary
means of obtaining a definitive diagnosis, using muscle pathology
and biochemistry to assist with interpretation of inconclusive genetic
results.
Recent evidence has provided important insights into the importance of nuclear-mtDNA genomic cross-talk and has provided a
descriptive framework for classifying and understanding disorders that
emanate from perturbations in this cross-talk. Although not strictly
considered as mtDNA genetic disorders, manifestations do overlap
those highlighted above (Fig. 468-10).
IMPACT OF HOMOPLASMIC SEQUENCE
VARIATION ON HERITABLE TRAITS AND
DISEASE
The relationship among the degree of heteroplasmy, tissue distribution
of the mutant mtDNA, and disease phenotype simplifies inference of
a clear causative relationship between heteroplasmic mutation and
disease. With the exception of certain mutations (e.g., those causing
most cases of LHON), drift to homoplasmy of such mutations would be
precluded normally by the severity of impaired oxidative phosphorylation and the consequent reduction in reproductive fitness. Therefore,
sequence variants that have reached homoplasmy should be neutral in
terms of human evolution and, hence, useful only for tracing human
evolution, demography, and migration, as described above. Thus, novel
homoplasmic variants are seldom pathogenic. One important exception is in the case of one or more of the homoplasmic population-level
variants, which designate the mtDNA haplogroup J, and the interaction with the mtDNA mutations causing LHON. Reduced disease
predilection suggests that one or more of the ancient sequence variants
designating mtDNA haplogroup J appear to attenuate predisposition to
degenerative disease, in the presence of other risk factors. Whether or
not additional epistatic interactions between population-level mtDNA
haplotypes and common health conditions will be found remains to
be determined. If such influences do exist, then they are more likely
to be relevant to health conditions in the postreproductive age groups,
wherein evolutionary filters would not have had the opportunity to
censor deleterious effects and interactions and wherein the effects of
oxidative stress during aging or with poor diet or lack of exercise may
play a role. Although much has been written about the possible associations between population-level common mtDNA variants and human
health and disease phenotypes or adaptation to different environmental influences (e.g., climate), a word of caution is in order.
Many studies that purport to show such associations with phenotypes such as longevity, athletic performance, and metabolic and
neurodegenerative disease are limited by small sample sizes, possible
genotyping inaccuracies, and the possibility of population stratification
or ethnic ancestry bias. Because mtDNA haplogroups are so prominently partitioned along phylogeographic lines, it is difficult to exclude
the possibility that a haplogroup for which an association has been
reported is simply a marker for otherwise unappreciated population
Clinical investigations
Blood: creatine kinase, liver functions,
glucose, lactate
Urine: organic and amino acids
CSF: glucose, protein, lactate
Cardiac x-ray, ECG, ECHO
EEG, EMG, nerve conduction
Brain CT/MRI
PCR/RFLP analysis of
blood for known mutations
Histochemistry Study of respiratorychain complexes
activities
Yes
No
Molecular genetic analysis
rearrangements
PCR/RFLP for common point mutation
mtDNA automated sequencing
Specific point mutation syndrome:
e.g., MELAS, MERRF, and LHON
Muscle biopsy
FIGURE 468-9 Clinical and laboratory investigation of a suspected mitochondrial
DNA (mtDNA) disorder. CSF, cerebrospinal fluid; CT, computed tomography; ECG,
electrocardiogram; ECHO, echocardiography; EEG, electroencephalogram; EMG,
electromyogram; LHON, Leber’s hereditary optic neuropathy; MELAS, mitochondrial
encephalomyopathy, lactic acidosis, and stoke-like episodes; MERRF, myoclonic
epilepsy with ragged red fibers; MRI, magnetic resonance imaging; PCR, polymerase
chain reaction; RFLP, restriction fragment length polymorphism.
Mitochondrial DNA and Heritable Traits and Diseases
3677CHAPTER 468
Multiple ∆mtDNA
dNTP pool
Pyrimidine salvage
mtDNA depletion
adPEO
Twinkle
Twinkle
Alpers’ like
IOSCA
Alpers’s.
SCAE
Pol γA
Pol γ
adPEO
arPEO
A
B
ANT1
adPEO
Deoxyguanosine kinase
MPV17
Thymidine kinase (TK2)
RRMB2 (p53-R2)
Succinyl-CoA synthase
(SUCLA2, SUCLG1)
TP
Thymidine phosphorylase
Patient Control
FIGURE 468-10 Disorders associated with perturbations in nuclear-mitochondrial genomic crosstalk. Clinical features and genes associated with multiple mitochondrial DNA (mtDNA) deletions,
mtDNA depletion, and mitochondrial neurogastrointestinal encephalomyopathy syndromes.
ANT, adenine nucleotide translocators; adPEO, autosomal dominant progressive external
ophthalmoplegia; arPEO, autosomal recessive progressive external ophthalmoplegia; IOSCA,
infantile-onset spinocerebellar ataxia; SCAE, spinocerebellar ataxia and epilepsy. (Reproduced with
permission from A Spinazzola, M Zeviani: Disorders from perturbations of nuclear-mitochondrial
intergenomic cross-talk. J Intern Med 265:174, 2009.)
tissue is expected to be associated with mitochondrial
dysfunction, as reflected in an age-associated patchy
and reduced COX activity on histochemical staining,
especially in skeletal and cardiac muscle and brain.
A particularly well-studied and potentially important
example is the accumulation of mtDNA deletions and
COX deficiency observed in neurons of the substantia
nigra in Parkinson’s disease patients.
The progressive accumulation of ROS has been
proposed as the key factor connecting mtDNA mutations with aging and age-related disease pathogenesis
(Fig. 468-11). As noted above, ROS are a by-product
of normal oxidative phosphorylation and are removed
by detoxifying antioxidants into less harmful moieties;
however, environmental factors or mutations that result
in exaggerated production of ROS or impaired removal
result in ROS accumulation and subsequent cellular
injury. One of the main targets for ROS-mediated injury is
DNA, and mtDNA is particularly vulnerable because of its
proximity to the origin of free radical production, the lack
of protective histones, and less efficient injury repair systems compared with nuclear DNA. In turn, accumulation
of mtDNA mutations results in inefficient oxidative phosphorylation, with the potential for excessive production of
ROS, generating a “vicious cycle” of cumulative mtDNA
damage. Indeed, measurement of the oxidative stress
biomarker 8-hydroxy-2-deoxyguanosine has been used
to measure age-dependent increases in mtDNA oxidative
damage at a rate exceeding that of nuclear DNA. It should
be noted that mtDNA mutations can potentially occur in postmitotic
cells as well, because mtDNA replication is not synchronized with the
cell cycle. Two other proposed links between mtDNA mutation and
aging, besides ROS-mediated tissue injury, are the perturbations in
efficiency of oxidative phosphorylation with disturbed cellular aerobic
function and perturbations in apoptotic pathways, whose execution
steps involve mitochondrial activity.
heterogeneity, wherein a nongenetic (societal or environmental) difference among the populations marked by the mtDNA haplogroup
differences is actually causally related to the disease of interest. The
experimental difficulty in generating cellular or animal models to test
the functional influence of homoplasmic sequence variants (as a result of
mtDNA polyploidy) further compounds the challenge. The most likely
formulation is that the risk conferred by different mtDNA haplogroupdefining homoplasmic mutations for common diseases depends on the
concomitant nuclear genomic background, together with environmental influences. Progress in minimizing potentially misleading associations in mtDNA heritable trait and disease studies should include
ensuring adequate sample size taken from a large sample recruitment
base, using carefully matched controls and population structure determination, and performing analysis that takes into account epistatic
interactions with other genomic loci and environmental factors.
IMPACT OF ACQUIRED SOMATIC mtDNA
MUTATION ON HUMAN HEALTH AND
DISEASE
Studies on aging humans and animals have shown a potentially
important correlation of age with the accumulation of heterogeneous
mtDNA mutations, especially in organ systems that undergo the most
prominent age-related degenerative tissue phenotype. Sequencing of
PCR-amplified single mtDNA molecules has demonstrated an average
of two to three point mutations per molecule in elderly subjects when
compared with younger ones. Point mutations observed include those
responsible for known heritable heteroplasmic mtDNA disorders, such
as the m.3344A>G and m.3243A>G mutations responsible for the
MERRF and MELAS syndromes, respectively. However, the cumulative burden of these acquired somatic point mutations with age was
observed to remain well below the threshold expected for phenotypic
expression (<2%). Point mutations at other sites not normally involved
in inherited mtDNA disorders have also been shown to accumulate
to much higher levels in some tissues of elderly individuals, with
the description of tissue-specific “hot spots” for acquired somatic
mtDNA point mutations. Likewise, an age-associated and tissuespecific accumulation of mtDNA deletions has been observed, including deletions involved in known heritable mtDNA disorders, as well as
others. The accumulation of functional mtDNA deletions in a given
Mutant
mitochondrial
proteins
Damaged
mitochondrial
proteins
Error-prone
DNA Pol-γ
Nuclear
DNA damage
ROS
Decreased
DNA repair
Apoptosis
DNA
mutations
O2
O2
H2O2
H2O OH
Aging
X − X
FIGURE 468-11 Multiple pathways of mitochondrial DNA (mtDNA) damage and
aging. Multiple factors may impinge on the integrity of mitochondria that lead to
loss of cell function, apoptosis, and aging. The classic pathway is indicated with
blue arrows; the generation of reactive oxygen species (ROS; superoxide anion,
hydrogen peroxide, and hydroxyl radicals), as a by-product of mitochondrial
oxidative phosphorylation, results in damage to mitochondrial macromolecules,
including the mtDNA, with the latter leading to deleterious mutations. When these
factors damage the mitochondrial energy-generating apparatus beyond a functional
threshold, proteins are released from the mitochondria that activate the caspase
pathway, leading to apoptosis, cell death, and aging. (Reproduced with permission
from L Loeb et al: The mitochondrial theory of aging and its relationship to reactive
oxygen species damage and somatic mtDNA mutations. Proc Natl Acad Sci USA
102:18769, 2005. Copyright (2005) National Academy of Sciences, U.S.A.)
3678 PART 16 Genes, the Environment, and Disease
Genetic intervention studies in animal models have sought to
clarify the potential causative relationship between acquired somatic
mtDNA mutation and the aging phenotype and the role of ROS in
particular. Replication of the mitochondrial genome is mediated by
the activity of the nuclear-encoded POLG. A transgenic homozygous
mouse knock-in mutation of this gene renders the polymerase enzyme
deficient in proofreading and results in a threefold to fivefold increase
in mtDNA mutation rate. Such mice develop a premature aging phenotype, which includes subcutaneous lipoatrophy, alopecia, kyphonia, and weight loss with premature death. Although the finding of
increased mtDNA mutation and mitochondrial dysfunction with age
has been solidly established, the causative role and specific contribution of mitochondrial ROS to aging and age-related disease in humans
have yet to be proved. Similarly, although many tumors display higher
levels of heterogeneous mtDNA mutations, a causal relationship to
tumorigenesis has not been proved.
Besides the age-dependent acquired accumulation in somatic cells
of heterogeneous point mutations and deletions, a quite different effect
of nonheritable and acquired mtDNA mutations has been described
affecting tissue stem cells. In particular, disease phenotypes attributed
to acquired mtDNA mutation have been observed in sporadic and
apparently nonfamilial cases involving a single individual or even tissue,
usually skeletal muscle. The presentation consists of decreased exercise
tolerance and myalgias, sometimes progressing to rhabdomyolysis. As
in the case of the sporadic, heteroplasmic, large-scale deletion, classic
syndromes of chronic PEO, Pearson’s syndrome, and KSS, the absence of
a maternal inheritance pattern and the finding of limited tissue distribution suggest a molecular pathogenic mechanism emanating from mutations arising de novo in muscle stem cells after germline differentiation
(somatic mutations that are not sporadic and occur in tissue-specific
stem cells during fetal development or in the postnatal maintenance or
postinjury repair stage). Such mutations would be expected to be propagated only within the progeny of that stem cell and affect a singular
tissue within a given individual, without evidence of heritability.
PROSPECTS FOR CLINICAL MANAGEMENT
OF mtDNA DISEASE
■ TREATMENT OF mtDNA DISORDERS
No specific curative treatment for mtDNA disorders is currently available; therefore, the management of mitochondrial disease is largely supportive. Management issues may include early diagnosis and medical
management of epilepsy, gastrointestinal dysfunction, weakness, diabetes mellitus, cardiac dysrhythmia, hearing loss, endocrinopathy, ptosis,
and cataracts. The value of aggressive symptom management cannot be
understated. Less specific interventions in the case of other disorders
involve combined treatment strategies including dietary intervention
and removal of toxic metabolites. Cofactors and vitamin supplements
are widely used in the treatment of diseases of mitochondrial oxidative
phosphorylation, although there is little evidence, apart from anecdotal
reports, to support their use. This includes administration of artificial
electron acceptors, including vitamin K3
, vitamin C, and ubiquinone
(CoQ10); administration of cofactors (coenzymes) including riboflavin,
carnitine, and creatine; and use of oxygen radical scavengers, such as
vitamin E, copper, selenium, ubiquinone, and idebenone. Drugs that
could interfere with mitochondrial function, such as the anesthetic
agent propofol, barbiturates, and high doses of valproate, should be
avoided. The use of valproate in patients with pathogenic mutations in
POLG and possibly other mutations affecting mtDNA stability and replication is especially contraindicated. Supplementation with the nitric
oxide synthase substrate l-arginine and, more recently, l-citrulline has
been advocated as a vasodilator treatment during stroke-like episodes
as well as for chronic management in patients with MELAS. Open-label
studies demonstrate that levoarginine and levocitrulline may be helpful
in reducing the stroke-like symptoms in MELAS but may have serious
side effects. As CSF folate deficiency has been reported in some cases of
mitochondrial disease, this can be treated with folinic acid.
The physician should also be familiar with environmental interactions, such as the strong and consistent association between visual loss
in LHON and smoking or ethanol consumption. A clinical penetrance
of 93% was found in men who smoked. Asymptomatic carriers of an
LHON mtDNA mutation should, therefore, be strongly advised not
to smoke and to moderate their alcohol intake. Although not a cure,
these interventions might stave off the devastating clinical manifestations of the LHON mutation. Another example is strict avoidance of
aminoglycosides in the familial syndrome of ototoxic susceptibility to
aminoglycosides in the presence of the mtDNA m.1555A>G mutation
of the 12SrRNA encoding gene.
Clinical trials using novel agents have been initiated and launched.
These agents include α-tocotrienol (PTC-743 previously termed EPI743, PTC Therapeutics), REN-001 (Reneo Pharmaceuticals), ASP0367
(Astellas Pharmaceuticals), and elamipretide (Stealth Biotherapeutics).
In an open-label study of α-tocotrienol used to treat 10 children with
Leigh’s syndrome, there were improvements in the primary endpoints,
including the Newcastle Pediatric Mitochondrial Diseases Scale, the
Gross Motor Function Measure, and the PedsQL Neuromuscular
Module. Ongoing studies continue for α-tocotrienol in children with
epilepsy, along with REN001, ASP0367, and elamipretide in adults
with primary mitochondrial myopathy. Conclusive progress has experienced some delay due to the COVID-19 pandemic.
GENETIC COUNSELING, PRENATAL
DIAGNOSIS, AND PGD IN mtDNA
DISORDERS
The provision of accurate genetic counseling and reproductive options
to families with mtDNA mutations is challenging due to the unique
genetic features of mtDNA inheritance that distinguish it from Mendelian genetics. mtDNA defects are transmitted by maternal inheritance.
mtDNA de novo mutations are often large deletions, affect one family
member, and usually represent no significant risk to other members of
the family. In contrast, mtDNA point mutations or duplications can be
transmitted maternally. Accordingly, the father of an affected individual has no risk of harboring the disease-causing mutation, and a male
cannot transmit the mtDNA mutation to his offspring. In contrast, the
mother of an affected individual usually harbors the same mutation but
may be completely asymptomatic. This wide phenotypic variability is
primarily related to the phenomena of heteroplasmy and the mutation
load carried by different members of the same family. Consequently,
a symptomatic or asymptomatic female harboring a disease-causing
mutation in a heteroplasmic state will transmit to her offspring variable amounts of the mutant mtDNA molecules. The offspring will be
symptomatic or asymptomatic primarily according to the mutant load
transmitted via the oocyte and, to some extent, subsequent mitotic
segregation during development. Interactions with the mtDNA haplotype background or nuclear human genome (as in the case of LHON)
serve as an additional important determinant of disease penetrance.
Because the severity of the disease phenotype associated with the
heteroplasmic mutation load is a function of the stochastic differential
segregation and copy number of mutant mtDNA during the oogenesis
bottleneck and, subsequently, following tissue and organ development
in the offspring, it is rarely predictable with any degree of accuracy.
For this reason, prenatal diagnosis (PND) and PGD techniques that
have evolved into integral and well-accepted standards of practice are
severely hampered in the case of mtDNA-related diseases.
The value of PND and PGD is limited, partly due to the absence
of data on the rules that govern the segregation of wild-type and
mutant mtDNA species (heteroplasmy) among tissue in the developing
embryo. Three factors are required to ensure the reliability of PND and
PGD: (1) a close correlation between the mutant load and the disease
severity, (2) a uniform distribution of mutant load among tissues, and
(3) no major change in mutant load with time. These criteria are suggested to be fulfilled for the NARP m.8993T>G mutation but do not
seem to apply to other mtDNA disorders. In fact, the level of mutant
mtDNA in a chorionic villous or amniotic fluid sample may be very
different from the level in the fetus, and it would be difficult to deduce
whether the mutational load in the prenatal samples provides clinically
useful information regarding the postnatal and adult state.
Mitochondrial DNA and Heritable Traits and Diseases
3679CHAPTER 468
■ PREVENTION OF MITOCHONDRIAL DISEASE
INHERITANCE BY ASSISTED REPRODUCTIVE
TECHNOLOGIES
Because the treatment options for patients with mitochondrial disease
are rather limited, with no current U.S. Food and Drug Administration (FDA)–approved therapies for established mitochondrial DNA
disease, preventive interventions that eliminate the likelihood of
transmission of affected mtDNA into offspring are desirable. The poor
reliability of prenatal and preimplantation approaches in predicting
mitochondrial DNA disease has resulted in the search for alternative
preventive approaches. The common purpose underlying various
emerging approaches is to reduce mutant heteroplasmy levels to a
level below a pathogenic threshold. This is based on the observed
relationship between heteroplasmy and disease inheritance patterns,
which indicates that even a small increase in copy number of nonmutant mtDNA molecules in the fertilized egg can exceed the threshold
required to ameliorate serious clinical disease. Use of gene editing, with
clustered regularly interspaced short palindromic repeats (CRISPR) or
mitochondrial-targeted TALEN (transcription activator-like effector
nucleases) technology, for example, to shift the heteroplasmy load
in affected tissues will require future development of corrective gene
delivery techniques. Likewise, induced pluripotent cell technology has
not yet met with widespread success in the preclinical research setting.
This has prompted the application of mitochondrial replacement therapy (MRT) approaches (Fig. 468-12). These approaches substitute in
vitro the entire oocyte or zygote complement of mitochondria, together
FIGURE 468-12 Mitochondrial replacement techniques—maternal spindle transfer and pronuclear transfer. In both procedures, some mutant mtDNA, estimated at 1–2%,
might be carried over together with the spindle or pronucleus, but the levels are low enough to avoid disease risk. IVF, in vitro fertilization. (From MJ Falk et al: Mitochondrial
replacement techniques—Implications for the clinical community. N Engl J Med 374:1103, 2016. Copyright © 2016, Massachusetts Medical Society. Reprinted with
permission from Massachusetts Medical Society.)
3680 PART 16 Genes, the Environment, and Disease
■ DEFINITION
In telomere diseases (also called telomeropathies or telomere spectrum
disorders), organ dysfunction is caused by loss of the ends of chromosomes, a process termed accelerated telomere attrition. Inadequate
repair or insufficient protection of telomeres and their resulting erosion induces cell death, deficient cell proliferation, and chromosome
instability; affected tissues show defective organ regeneration, fibrosis
469 Telomere Disease
Rodrigo T. Calado, Neal S. Young
with their mtDNA from the carrier mother, with the unaffected complement of mitochondria and their unaffected mtDNA from a donor
woman. This can be accomplished either by removing and transferring
the carrier mother’s spindle with her nuclear DNA into the unfertilized
oocyte of the donor or, alternatively, by transferring the pronucleus
from the fertilized oocyte of the carrier mother to the unfertilized
donor oocyte from which the pronucleus has been removed. These
approaches provide a “bulk” substitution and hence do not target the
specific mtDNA mutation, and they are potentially applicable to a wide
variety of mtDNA disorders. This is a form of germline genetic therapy,
and therefore, it projects onto future generations in the case of a female
offspring. Accordingly, ethical and regulatory bodies have appropriately weighed in on the societal implications of such approaches and
have been tentatively supportive of human clinical investigation for
situations that would prevent great suffering and when the clinical
need is clear and unambiguous, subject to specified conditions and
principles and subject to ethical scrutiny. Several such studies have
been initiated, and careful examination and follow-up are needed to
determine developmental and longer-term health and fertility of children who have undergone genetic manipulation at the earliest stages of
human development and whose genomes comprise separate maternal
origins of nuclear and mtDNA genomes. It has been recommended
that such studies be limited to male offspring, who cannot then transmit the donor mtDNA to future generations, until such time as the
health, ethical, and societal issues are well understood and live up to
the exciting promise of reducing the burden of clinical mtDNA disease
in the future.
■ FURTHER READING
Alston CL et al: The genetics and pathology of mitochondrial disease.
J Pathol 241:236, 2017.
Camp K et al: Nutritional interventions in primary mitochondrial
disorders: Developing an evidence base. Mol Genet Metab 119:187,
2016.
DiMauro S: Mitochondrial encephalomyopathies—Fifty years on: The
Robert Wartenberg Lecture. Neurology 81:281, 2013.
DiMauro S et al: The clinical maze of mitochondrial neurology. Nat
Rev Neurol 9:429, 2013.
Haas RH et al: Mitochondrial disease: A practical approach for primary care physicians. Pediatrics 129:1326, 2007.
Koopman WJ et al: Monogenic mitochondrial disorders. N Engl J Med
366:1132, 2012.
Parikh S et al: Patient care standards for primary mitochondrial
disease: A consensus statement from the Mitochondrial Medicine
Society. Genet Med 19:1380, 2017.
Russell OM et al: Mitochondrial diseases: Hope for the future. Cell
181:168, 2020.
Saneto RP et al (eds): Mitochondrial Case Studies, Underlying Mechanisms and Diagnosis. London, Academic Press/Elsevier, 2016.
or replacement by fat, and a proclivity for cancer. A variety of regenerative disorders affecting especially the bone marrow, lungs, liver, and
skin share telomere dysfunction and accelerated loss as their common
molecular mechanism. Cross-sectional data of average telomere length
in cohorts of different ages and longitudinal studies in individuals indicate that telomeres shorten with aging at an average of 50 base pairs/
year in peripheral blood leukocytes. Despite shortening of telomeres
over time, normal aging is not associated with the development of a
telomere disease from short telomeres. In normal aging, sufficient stem
cell number and function are maintained to sustain vital processes.
For example, in stem cell transplant, even a limited number of donor
hematopoietic cells maintains normal hematopoiesis for many years, at
least in part related to normal telomerase function and telomere repair.
Telomere length associates with life span in the general population.
While shorter leukocyte telomeres correlate with increased mortality
risk, especially from nonmalignant causes, telomere loss is not established as the cause of physiologic aging.
■ DISEASE MECHANISM
Telomeres, the physical termini of linear chromosomes, are repeated
hexanucleotide sequences physically associated with specific proteins.
Telomeres function to protect the chromosome ends against recognition as damaged or infectious DNA by the DNA repair machinery
(Fig. 469-1). During mitosis, the DNA polymerase employs an RNA
oligonucleotide with a 3′ hydroxyl group to prime replication. The
primer dissociates as the DNA polymerase advances along the template strand, and a gap is left at the ends of linear DNA molecules: the
newly synthesized DNA strand is necessarily shorter than the original
template—the “end-replication problem.” Chromosome erosion is thus
inevitable with mitotic cell division, but the noncoding telomeric long,
repetitive structure buffers loss of genetic information. In human cells,
telomeres are composed of hundreds to thousands of TTAGGG tandem
repeats in the leading and CCCTAA in the lagging DNA strand. At
birth, telomeres are relatively long, but they inexorably shorten with
chronological aging (Fig. 469-1). In an individual cell, critically short
telomere length triggers the p53 pathway, usually leading to proliferative
arrest, senescence, and apoptosis. Telomere loss is the molecular basis
for the “Hayflick phenomenon,” the limit to cell division and thus to cell
proliferation in tissue culture. If a cell overcomes proliferation arrest,
extremely short telomeres engage the DNA damage repair machinery,
and chromosome end-to-end fusions, chromosome breaks, aneuploidy,
and chromosome instability may occur. In addition to the telomere
repeated sequences, a group of specialized proteins, collectively termed
shelterins, directly bind to or indirectly associate with telomeres, assisting in the organization of the telomere tertiary structure and inhibiting
activity of DNA damage response proteins (Fig. 469-1).
To escape telomere attrition, cells with high proliferative demand,
including embryonic and adult stem cells, lymphocytes, and the
majority of cancer cells have at least two mechanisms to preserve
telomere length: homologous recombination (important in malignant cell transformation) and the synthesis of telomeric repeats (in
physiologic telomere maintenance). The telomerase machinery adds
GTTAGG hexanucleotides to the 5′ end of the leading DNA strand
using telomerase, a reverse transcriptase enzyme (TERT), and TERC,
its RNA template (Fig. 469-1). The telomerase holoenzyme complex is
composed of two copies of TERT, TERC, and dyskerin, and associated
proteins. TERC binds to TERT and serves as the RNA template for its
function as a reverse transcriptase. Dyskerin, encoded by DKC1, stabilizes the complex, and TCAB1, encoded by the WRAP53 gene, aids
telomerase trafficking to the Cajal bodies, nuclear structures for ribonucleoprotein processing where telomerase associates with telomeres
for elongation. Telomerase expression is tightly regulated. MYC, sex
hormones, and many additional factors stimulate TERT transcription.
In mature cells, the TERT gene is repressed. In addition, shelterin proteins can also regulate telomerase function and processivity, modulating its catalytic activity on telomeres. Other proteins also are important
for telomere length maintenance. RTEL1, an essential DNA helicase,
dismantles T-loops and resolves G-quadruplexes, ensuring adequate
telomere elongation.
Telomere Disease
3681CHAPTER 469
When telomeres are critically short, the DNA damage response machinery may be recruited, mistaking
telomeres for damaged or infectious DNA and forcing
inappropriate repair. Activation of this pathway may
cause chromosome instability due to end-to-end fusion
of chromosomes or translocations; these alterations
generate genomic instability and potentially malignant
clones of cells. That telomere dysfunction increases the
risk of cancer development has been demonstrated in
murine models of telomerase deficiency, and patients
with telomere diseases are prone to develop acute
myeloid leukemia and head and neck squamous cell
carcinomas.
■ GENETICS
The pattern of inheritance is variable: X-linked, autosomal recessive, and autosomal dominant. Complex
genetic architecture also occurs, and affected patients
may inherit pathogenic loss-of-function variants in
more than one gene involved in the same telomere biology pathway. At least 15 genes have been implicated in
the etiology of telomeropathies (Table 469-1).
■ CLINICAL MANIFESTATIONS
Presentation of telomere disease in the clinic is highly
variable—in the tissues affected, in the severity of organ
dysfunction, and in patterns of disease within families
and between families with similar mutations. In a
same family, one individual may be severely affected,
but close relatives carrying the same mutation may
be asymptomatic and with normal laboratory results.
Asymptomatic carriers, however, may have subclinical
organ dysfunction, which may be detected by directed
or specialized testing (reduced forced vital capacity on
pulmonary function test, hypocellular bone marrow
at biopsy, hepatic steatosis on ultrasound). Somatic
genetic rescue may occur in telomere diseases and
modulate the clinical phenotype. Rescue is a rare
spontaneous genetic event in a somatic cell, conferring
a selective advantage and annulling the effect of the
original pathogenic germline mutation. In telomeropathies, somatic genetic rescue has been detected in
hematopoietic cells, so that one clonal population
adequately produces blood cells and mitigates the marrow failure phenotype. Generally, relatives sharing the
same mutation and short telomeres may have different
organs affected: for instance, while one individual may
have aplastic anemia, another within the pedigree may
suffer from pulmonary fibrosis. Environmental regenerative stresses, factors such as smoking and alcohol consumption,
and viral infection may increase susceptibility to organ damage in
these patients and contribute to disease heterogeneity.
Disease anticipation, in which clinical phenotype manifests at an
earlier age in successive generations, is observed in some families with
telomeropathies, due to the direct inheritance of short telomeres present in sperm and oocytes.
The diagnosis of a telomere disease is suggested by personal and
family history, strengthened by simple measurement of leukocyte telomere length, and usually definitively established by next-generation
sequencing for genes encoding telomere repair enzyme complex and
shelterin components.
Dyskeratosis Congenita Dyskeratosis congenita is the classic
telomere disease, a mainly pediatric syndrome diagnosed in the first
two decades of life. Affected children often have at least two features
of the mucocutaneous triad of ungual dystrophy, reticular skin pigmentation, and oral leukoplakia (Fig. 469-2). In severe syndromes,
affected newborns have cerebellar hypoplasia (Hoyeraal-Hreidarsson
B Age (years)
A
0
Telomere length (kb)
2
4
6
8
10
12
14
Shelterin
TRF2
Rap1
TPP1
TIN2 TRF1
POT1
Dyskerin
AAUCCC
Telomerase
AATCCCAATCCCAATCCC-5'
GAR
NOP10
NHP2
RNA
template
Centromere
Chromosome
T loop
D loop
3'
5'
TERT
Average loss of 40–60 base pairs/year
0 20 40 60 80 100
TTAGGGTTAGGGTTAGGGTTAGGGTTAGGG-3'
FIGURE 469-1 Telomeres and telomerase. A. Telomeres are ribonucleoprotein structures located at
the termini of linear chromosomes inside the cell nucleus composed of hundreds of tandem hexameric
DNA repeats. A group of proteins bind directly or indirectly to telomere sequences in order to provide
protection to the structure and are collectively termed shelterin or telosome (TRF1, TRF2, TIN2, POT1,
TPP1, and RAP1). As the 3′ end of the leading strand forms a single-stranded overhang, it folds back and
invades the telomeric double helix, forming a lariat termed T-loop. The telomerase complex is composed
of the enzyme telomerase reverse transcriptase (TERT), its RNA component (TERC), the protein dyskerin,
and associated proteins (NHP2, NOP10, and GAR). This enzymatic complex elongates telomeres by
adding GTTAGG hexameric repeats to the 3′ end of the telomeric leading strand, using a sequence in
TERC as the template. B. The average telomere length in human leukocytes varies: it is longer at birth
(10–11 kb) and progressively shortens with aging (5–7 kb at age 90 years) at an average loss of 40–60
base pairs/year. However, there is significant variability in telomere length in each given age.
Pathologic accelerated telomere attrition has a genetic origin.
Germline loss-of-function mutations in genes involved in telomere
maintenance and function impair telomere length repair, thus increasing the rate of telomere erosion in highly proliferative cells. Telomeres
reach critically short lengths faster than with normal aging; the consequences are limited cell proliferation and impaired tissue regeneration.
Some organs appear to be particularly susceptible to telomere erosion.
Billions of blood cells are produced daily (Chap. 96), and telomere
attrition curtails cell proliferation, producing a hypocellular marrow
and often low blood counts. The liver also is an organ with high proliferative capacity, and telomere dysfunction impairs hepatic regeneration
after injury, with a variety of pathologic consequences. The lung alveolar epithelium is in contact with exogenous toxins that stimulate regeneration, and telomere loss may hinder these physiologic responses.
However, it remains unclear why other regenerative tissues, such as the
intestine, are less affected by telomere dysfunction; the mechanism by
which telomere loss provokes a fibrotic response in the lungs (pulmonary fibrosis), an adipose response in the marrow (aplastic anemia),
and both in the liver (hepatic steatosis and cirrhosis) is also unclear.
3682 PART 16 Genes, the Environment, and Disease
TABLE 469-1 Genetic Variants in 13 Genes Involved in Telomere Maintenance, Inheritance
Pattern, and Phenotype
GENE
DYSKERATOSIS
CONGENITA
APLASTIC
ANEMIA
PULMONARY
FIBROSIS CIRRHOSIS MDS/LEUKEMIA
Telomerase
DKC1 XL
TERT AD/AR AD/AR AD AD AD/AR
TERC AD/AR AD AD AD AD
NOP10 AR
NHP2 AR
WRAP53 AR
Shelterin
TINF2 AD AD AD
TERF2 AD
ACD AD
Others
RTEL1 AR AD/AR AD AD
CTC1 AR AR
PARN AD
USB1 AD
ZCCHC8 AD AD
NAF1 AD
Abbreviations: AD, autosomal dominant; AR, autosomal recessive; MDS, myelodysplastic syndrome; XL, X-linked.
FIGURE 469-2 Skin manifestations of dyskeratosis congenita. The pediatric syndrome dyskeratosis congenita
is characterized by the mucocutaneous triad of (A) reticular skin pigmentation, (B) oral leukoplakia, and (C, D) nail
dystrophy.
syndrome) or exudative retinopathy (Revesz syndrome) (Fig. 469-3).
Telomeres are usually extremely short, below the first percentile
expected for age (Fig. 469-4). Most patients with dyskeratosis congenita develop bone marrow failure, often requiring transfusions and
ultimately bone marrow transplant. Pulmonary fibrosis appears in as
many as 20% of cases and liver disease in 10%, often after bone marrow
transplant for hematopoietic failure. Other tissues and organs also may
be affected (Fig. 469-3). The most common mutations in dyskeratosis
congenita patients are in the DKC1, TINF2, TERT, TERC, and RTEL1
genes, and triallelic inheritance (involving two genes in the same pathway) also may occur (Table 469-1).
Bone Marrow Failure Aplastic anemia
(Chap. 102) is the most common major clinical
manifestation of dyskeratosis congenita. However, young or older patients carrying a telomere defect, without typical physical stigmata
of dyskeratosis, also may also develop marrow
failure. Genetic variants usually are monoallelic
(one mutated allele and one wild-type allele),
and TERT, TERC, and RTEL1 are the genes
usually affected. Telomere loss in these cases
is often less intense than in classic dyskeratosis
congenita. As a result of inadequate telomerase
function, the stem cell pool is limited in size
and in its ability to regenerate. There is insufficient production of erythrocytes, platelets,
and granulocytes with anemia, thrombocytopenia, and leukopenia of peripheral blood
and low marrow cellularity (Fig. 469-5). The
most typical presentation is moderate aplastic
anemia after a long history of macrocytic mild
to moderate anemia and/or thrombocytopenia,
with preservation of leukocyte numbers. A
comprehensive personal and family history is
important, querying especially for blood count
abnormalities and cytopenia as well as lung
and liver disease; early hair graying, while not
specific to telomeropathies, strongly suggests
telomere disease in the appropriate context.
Immunosuppressive therapy is generally
ineffective in these patients, and they may be more susceptible to
pulmonary or hepatic complications after hematopoietic stem cell
transplant.
Myeloid Neoplasms Some patients diagnosed with myelodysplastic syndrome (Chap. 102) or acute myeloid leukemia (Chap. 104) have
a family history of bone marrow failure or of other myeloid neoplasms.
One of the genetic causes for myeloid neoplasia predisposition is a
telomere defect, and these disorders are now classified together by the
World Health Organization as “myeloid neoplasms associated with
telomere biology disorders.” Telomere length measurement may be
confounded by the presence of circulating immature cells, which may have very
short telomeres, precluding appropriate
test interpretation.
Pulmonary Fibrosis Pulmonary
fibrosis appears in ~20% of children
with dyskeratosis congenita, and conversely, ~10–15% of patients with
idiopathic pulmonary fibrosis (Chap.
293) or familial pulmonary fibrosis have an etiologic telomerase gene
mutation. Most pulmonary fibrosis
patients, regardless of mutation status,
have short telomeres for their age but
not as short as in dyskeratosis congenita. How telomere erosion causes
pulmonary fibrosis is unclear, but it
might prevent adequate proliferation
and regeneration of pneumocytes type
II. Idiopathic pulmonary fibrosis due
to a telomere disease usually manifests
after the fourth decade of life, with a
restrictive pattern on pulmonary function testing associated with decreased
diffusion capacity for carbon monoxide (DLCO) and a diffuse “honeycomb”
appearance on high-resolution CT
Telomere Disease
3683CHAPTER 469
FIGURE 469-3 Clinical consequences of telomere diseases. Telomere dysfunction affects a variety of organs:
cerebellum, eyes, lungs, liver, skin, gastrointestinal tract, and the bone marrow.
(Fig. 469-5). Histopathology of biopsied lung most commonly
shows usual interstitial pneumonia. The pulmonary clinical presentation in telomere disease is indistinguishable from idiopathic
pulmonary fibrosis, except that those with an underlying telomere
defect may have cryptic hepatic cirrhosis, macrocytosis, cytopenias,
and a family history of lung, liver, or bone marrow disease. Pulmonary arteriovenous malformation leading to right-to-left shunting
is observed in patients with pulmonary fibrosis due to telomere
disease. Patients with idiopathic pulmonary fibrosis or familial pulmonary fibrosis should have leukocyte telomere length assayed and,
if telomeres are short, undergo screening for mutations in telomereassociated genes and telomeres; however, telomere length may be
normal in some cases despite the presence of pathogenic mutations.
TERT, TERC, RTEL1, and PARN are the most commonly mutated
genes.
Liver Disease Genetic telomere defects
may cause hepatic cirrhosis (Chap. 344),
nodular regenerative hyperplasia of the liver,
nonalcoholic fatty liver disease (Chap. 343),
and hepatocellular carcinoma (Chap. 82).
Hepatocytes of most patients with cirrhosis
have very short telomeres. Eroded telomeres
limit hepatocyte proliferation, especially
upon chronic injury. Additionally, hepatocytes with short telomeres display abnormal
metabolic pattern and defective mitochondrial function. Abnormal liver pathology
may be uncovered incidentally during the
evaluation of telomeropathy patients with
aplastic anemia or pulmonary fibrosis, but
cirrhosis also may be the sole or most prominent clinical presentation of a telomere
defect. A minority of individuals with cirrhosis associated with virus B or C infection
or alcohol-associated liver disease may carry
a telomere-associated gene mutation. Liver
histopathology is variable, but cirrhosis is
usually associated with inflammation and
inflammatory cells (Fig. 469-5), increased
iron deposit, positivity for CD34 in sinusoid
endothelial cells, and widening of hepatocyte
plates. Defective telomere maintenance may
increase susceptibility of the liver to environmental challenges, such as alcohol and viruses, increasing the risk for
developing severe hepatic disease in mutation carriers.
■ TELOMERE LENGTH MEASUREMENT
Length of telomeres can be accurately measured in peripheral blood
leukocytes by commercial laboratories. Of several methods available,
flow–fluorescent in situ hybridization (FISH) and quantitative realtime polymerase chain reaction (qPCR) are most widely utilized. Both
methods have advantages and limitations and require high-quality
samples, usually fresh or freshly processed, as cell death and DNA degradation impact the accuracy of testing. Results are usually expressed
as leukocyte telomere length in kilobases. However, the interpretation
of length must account for patient age due to physiologic telomere loss.
A normal range for telomeres is available for each year of age, with
length longest at birth and shortening at 40–60 base pairs per year
(Fig. 469-1). For each age bracket, the percentile curves are calculated,
and a given patient’s test result is interpreted in the context of normal
age variation: telomeres below the tenth percentile for age are defined
as “short,” and telomeres below the first percentile are considered “very
short” (Fig. 469-4).
Telomeres may also be short in groups of patients with some chronic
conditions, such as cardiovascular disease or diabetes. However, in
these settings, telomere erosion is not assumed to be etiologic, but
rather a consequence of chronic inflammation; telomere testing is
not known to have clinical utility and is not recommended. Likewise,
telomere length tests have no known clinical utility in the assessment
of aging and longevity or as a basis for therapeutic interventions absent
a diagnosis of telomere disease.
Flow-FISH uses a fluorescent-labeled nucleotide probe specific for
telomere repeats in order to estimate telomere content in an individual
cell by flow cytometry. It has the advantage of determining telomere
length in individual cells and in leukocyte subpopulations (neutrophils,
lymphocytes, monocytes); lymphocyte telomere shortening is more
specific for telomere diseases than in other cells. However, flow-FISH
requires intact cells for analysis, which are not always available, and
neutrophils are susceptible to damage during processing, freezing, and
thawing.
qPCR utilizes telomere-binding modified primers to measure
telomere content in comparison to a housekeeping gene in the whole
leukocyte population and thus does not require intact cells. qPCR
Age, years
Telomere length, kb
0
0
2
4
6
8
10
12
14
16
10 20 30 40 50 60 70 80 90
1%
10%
50%
90%
99%
TINF2
DKC1
TERT
TERC
RTEL1
Other genes
Peripheral blood leukocyte’s telomere
length in telomere diseases
FIGURE 469-4 Telomere length measurement in dyskeratosis congenita. Telomeres
shorten with aging, and solid curves represent the percentiles for age in healthy
subjects. Telomeres are considered “short” when below the tenth percentile and
very short when below the first percentile. In patients with dyskeratosis congenita,
telomeres are usually below the first percentile, regardless of the gene lesion.
3684 PART 16 Genes, the Environment, and Disease
FIGURE 469-5 Pathologic manifestations of telomere diseases. A. In the bone marrow, telomere erosion predisposes to aplastic anemia, characterized by an empty
hematopoietic marrow replaced by fat (hematoxylin and eosin). B. In the liver, telomere attrition predisposes to cirrhosis (hematoxylin and eosin). C. Telomere shortening
may also result in nodular regenerative hyperplasia of the liver (reticulin stain). D. In the lungs, telomere dysfunction predisposes to pulmonary fibrosis mainly in the
subpleural regions, which may be detected by high-resolution CT scan.
provides an estimate of the average telomere length of a given sample
without determining telomere length in individual cells. Good DNA
quality is essential for adequate qPCR testing and automation or semiautomation required for clinical purposes, as variability in conditions
among batches may result in interassay variation.
■ GENETIC TESTING
When a patient with a suspected telomeropathy has short or very
short telomeres, genetic screening for mutations in genes involved in
telomere maintenance and biology is indicated (Table 469-1).
Genetic testing should be restricted to patients with suspected telomere
disorders. Sanger sequencing has been used for screening purposes but
has been replaced by next-generation sequencing, and for telomerase
complex and shelterin genes, commercial panels are available. Mutations may be biallelic or triallelic, involving two genes (especially in
dyskeratosis congenita), but in most cases of aplastic anemia, myelodysplastic syndrome, acute myeloid leukemia, idiopathic pulmonary
fibrosis, and hepatic cirrhosis, only one allele is mutated. Thus, it is
crucial to appropriately interpret genetic screening results, as several
rare singleton polymorphisms of unknown significance have been
identified in large cohorts of healthy individuals. In silico analysis,
mutation location, and functional studies are utilized before declaring
a mutation pathogenic.
Genetic counseling is necessary after screening, as the inheritance
pattern may be autosomal dominant, mutation penetrance is highly
variable, and phenotypes may be diverse even within a pedigree. Potential family stem cell donors must be screened before transplantation to
ensure that they do not have mutations.
TREATMENT
Telomere Disease
Patients with severe aplastic anemia due to telomere disease may
undergo allogeneic hematopoietic stem cell transplant when a
suitable donor is available. Treatment-related mortality may be
increased due to pulmonary and hepatic complications, for which
reduced-intensity conditioning regimens may be advantageous.
Lung transplant for pulmonary fibrosis is feasible but often not performed due to coexisting cytopenias, especially thrombocytopenia,
and other comorbidities. Additionally, patients with pulmonary
fibrosis associated with telomere disease have a poorer outcome
after lung transplant and with nontransplant therapies. Similarly,
there is no specific treatment for the liver in telomere disease;
liver transplant has been attempted in rare cases. Telomeropathy
patients should be advised to avoid toxins (metal dust, busulfan,
amiodarone), ionizing radiation, cigarette smoke, and alcohol as
possibly harmful.
Long-term therapy with androgens may mitigate telomere attrition and even elongate leukocyte telomere length in humans. In
research trials, danazol improved blood counts in marrow failure
patients and reduced transfusion requirement.
■ FURTHER READING
Blackburn EH et al: Human telomere biology: A contributory and
interactive factor in aging, disease risks, and protection. Science
350:1193, 2015.
Calado RT, Young NS: Telomere diseases. N Engl J Med 361:2353,
2009.
Collins J, Dokal I: Inherited bone marrow failure syndromes. Hematology 20:433, 2015.
Gruber HJ et al: Telomeres and age-relatd diseases. Biomedicines
9:1335, 2021.
Gutierrez-Rodrigues F et al: Pathogenic TERT promoter variants in
telomere diseases. Genet Med 21:1594, 2019.
Holohan B et al: Telomeropathies: An emerging spectrum disorder. J
Cell Biol 205:289, 2014.
Newton CA et al: Telomere-related lung fibrosis is diagnostically
heterogeneous but uniformly progressive. Eur Respir J 48:1710, 2016.
Savage SA, Bertuch AA: The genetics and clinical manifestations of
telomere biology disorders. Genet Med 12:753, 2010.
Swaminathan AC et al: Lung transplant outcomes in patients with
pulmonary fibrosis with telomere-related gene variants. Chest
156:477, 2019.
Townsley DM et al: Bone marrow failure and the telomeropathies.
Blood 124:2775, 2014.
Townsley DM et al: Danazol treatment for telomere diseases. N Engl
J Med 374:1922, 2016.
Gene- and Cell-Based Therapy in Clinical Medicine
3685CHAPTER 470
Gene therapy is a novel area of therapeutics in which the active agent
is a nucleic acid sequence rather than a protein or small molecule. One
of the most powerful concepts in modern molecular medicine, gene
therapy has the potential to address a host of diseases for which there
are currently no available treatments. Because delivery of naked DNA
or RNA to a cell is an inefficient process, most gene therapy is carried
out using a vector, or gene delivery vehicle. These vehicles have generally been engineered from viruses by deleting some or all of the viral
genome and replacing it with the therapeutic gene of interest under the
control of a suitable promoter, but nonviral delivery vehicles such as
lipid nanoparticles have also been used (Table 470-1). Gene therapy
strategies can thus be described in terms of three essential elements:
(1) a gene delivery vehicle; (2) a gene to be delivered, sometimes called
the transgene; and (3) a physiologically relevant target cell to which the
DNA or RNA is delivered. The series of steps in which the vector and
donated DNA or RNA enter the target cell and express the transgene
is referred to as transduction. Gene delivery can take place in vivo, in
which the vector is directly injected into the patient, or, in the case
of hematopoietic, immune, and some other target cells, ex vivo, with
removal of the target cells from the patient, followed by return of the
gene-modified autologous cells to the patient after manipulation in
the laboratory. The latter approach effectively combines gene transfer
techniques with cellular therapies (Chap. 484).
Currently approved gene therapies rely on transfer of DNA, but
investigational RNA-based therapies have gained ground rapidly, partly
as a key component of CRISPR-based gene editing (guide RNAs) and,
more recently, as the active agent in two SARS-CoV-2 vaccines. This
chapter will focus primarily on approved therapies (Table 470-2), with
some discussion of investigational therapies in late-phase development
and earlier trials of historical interest.
Clinical trials of gene therapy have been under way since 1990;
the first gene therapy product to be licensed in the United States or
Europe was approved in 2012 (see below). Given that vector-mediated
gene therapy is arguably one of the most complex therapeutics yet
developed, typically consisting of both a nucleic acid and a protein
470 Gene- and Cell-Based
Therapy in Clinical
Medicine
Katherine A. High, Marcela V. Maus
component, this time course from first clinical trial to licensed product
is noteworthy for being similar to those seen with other novel classes
of therapeutics, i.e., monoclonal antibodies. Thousands of subjects
have been enrolled in gene transfer studies or have received approved
products, and serious adverse events have been rare. Some of the initial
trials were characterized by an overabundance of optimism and a failure to be appropriately critical of preclinical studies in animals; in addition, it was in some contexts not fully appreciated that animal studies
are only a partial guide to safety profiles of products in humans (e.g.,
insertional mutagenesis and human immune responses to the vector).
Clinical experience and laboratory research led to a more nuanced
understanding of the risks and benefits of these new therapies and
more sophisticated selection of disease targets. Currently, gene therapies are being developed for a variety of disease entities (Table 470-3).
GENE TRANSFER FOR GENETIC DISEASE
Gene transfer strategies for genetic disease generally involve gene
addition therapy, an approach characterized by transfer of the missing
gene to a physiologically relevant target cell. However, other strategies
are possible, including supplying a truncated form of the gene with
comparable biologic activity (e.g., a gene encoding B domain-deleted
factor VIII for hemophilia A); supplying a gene that achieves a similar biologic effect through an alternative pathway (e.g., utrophin in
place of dystrophin for Duchenne’s muscular dystrophy); supplying an
antisense oligonucleotide to alter splicing of a transcript to generate
a functional protein (e.g., nusinersen in the treatment of spinal muscular atrophy); or downregulating a harmful effect through a small
interfering or short hairpin RNA. From a therapeutic standpoint,
gene therapies for genetic disease fall into two distinct categories: first,
they may provide treatment for diseases that have hitherto lacked any
pharmacologic therapies; or second, they may provide an alternative to
complex medical regimens that are frequently characterized by significant nonadherence due to the burden of treatment (e.g., monthly red
blood cell transfusions and iron chelation in transfusion-dependent β
thalassemia).
Gene therapy for genetic disease requires long-term expression of
the transgene. Two distinct strategies are available to achieve this goal:
one is to transduce stem cells with an integrating vector, so that all
progeny cells will carry the donated gene; and the other is to transduce
long-lived, postmitotic cells, such as skeletal muscle or neurons. In
the case of long-lived cells, integration into the target cell genome is
unnecessary. Instead, because the cells are nondividing, the donated
DNA, even if stabilized predominantly in an episomal form, will give
rise to expression for the life of the cell. This latter approach mitigates
risks related to integration and insertional mutagenesis.
TABLE 470-1 Characteristics of Commonly Used Gene Delivery Vehicles
VIRAL NONVIRAL
FEATURES RETROVIRAL LENTIVIRAL ADENOVIRAL AAV LIPID NANOPARTICLES
Viral genome RNA RNA DNA DNA N/A
Cell division requirement Yes G1
phase No No No
Packaging limitation 8 kb 8 kb 8–30 kb 5 kb 10 kb or more
Immune responses to vector Few Few Extensive Few Few
Genome integration Yes Yes Poor Poor May be used to package
either RNA or DNA
Long-term expression Yes Yes No Yes Transient for RNA
Main advantages Persistent gene transfer
in dividing cells
Persistent gene transfer in
transduced tissues; no history
of insertional oncogenesis;
can add desired promoter
Highly effective in
transducing various
tissues
Elicits few
inflammatory
responses,
nonpathogenic
Limited immune
responses against the
vector
Main disadvantages History of insertional
oncogenesis (occurred
in multiple cases); only
used ex vivo; gene
silencing
Might induce oncogenesis in
some cases (not yet observed);
only used ex vivo
Viral capsid elicits strong
immune responses
Limited packaging
capacity
Currently available
reagents predominantly
target the liver
Abbreviations: AAV, adeno-associated virus; N/A, not applicable.
No comments:
Post a Comment
اكتب تعليق حول الموضوع