Next Article in Journal
Osteogenic Potential of Human Dental Pulp Stem Cells (hDPSCs) Growing on Poly L-Lactide-Co-Caprolactone and Hyaluronic Acid (HYAFF-11TM) Scaffolds
Next Article in Special Issue
Digenic Inheritance in Rare Disorders and Mitochondrial Disease—Crossing the Frontier to a More Comprehensive Understanding of Etiology
Previous Article in Journal
New Insights via RNA Profiling of Formalin-Fixed Paraffin-Embedded Lung Tissue of Pulmonary Fibrosis Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Red Flags in Primary Mitochondrial Diseases: What Should We Recognize?

by
Federica Conti
1,†,
Serena Di Martino
1,†,
Filippo Drago
1,
Claudio Bucolo
1,2,
Vincenzo Micale
1,
Vincenzo Montano
3,
Gabriele Siciliano
3,
Michelangelo Mancuso
3,* and
Piervito Lopriore
3
1
Department of Biomedical and Biotechnological Science, School of Medicine, University of Catania, 95123 Catania, Italy
2
Center for Research in Ocular Pharmacology-CERFO, University of Catania, 95213 Catania, Italy
3
Neurological Institute, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
*
Author to whom correspondence should be addressed.
These authors equally contributed to the work.
Int. J. Mol. Sci. 2023, 24(23), 16746; https://doi.org/10.3390/ijms242316746
Submission received: 1 November 2023 / Revised: 22 November 2023 / Accepted: 23 November 2023 / Published: 25 November 2023
(This article belongs to the Special Issue Rare Diseases and Neuroscience)

Abstract

:
Primary mitochondrial diseases (PMDs) are complex group of metabolic disorders caused by genetically determined impairment of the mitochondrial oxidative phosphorylation (OXPHOS). The unique features of mitochondrial genetics and the pivotal role of mitochondria in cell biology explain the phenotypical heterogeneity of primary mitochondrial diseases and the resulting diagnostic challenges that follow. Some peculiar features (“red flags”) may indicate a primary mitochondrial disease, helping the physician to orient in this diagnostic maze. In this narrative review, we aimed to outline the features of the most common mitochondrial red flags offering a general overview on the topic that could help physicians to untangle mitochondrial medicine complexity.

1. Introduction

PMDs are the most frequent genetic metabolic disorders in humans, with a prevalence of approximately 1 in 4300 cases [1]. Pathogenic variants in all 37 mitochondrial DNA (mtDNA) genes and in more than 400 nuclear DNA genes have been associated with direct or indirect defects of the OXPHOS, which is the hallmark of these diseases [2]. These unique features of mitochondrial genetics and their ubiquitousness in human tissues explain PMDs’ vast clinical heterogeneity. PMDs are multisystemic diseases, mainly affecting high-energy demanding tissues, such as the muscles and central and peripheral nervous systems. Age of onset is variable, with the most severe defects generally appearing in childhood [3]. Cellular mtDNA polyplasmy, tissue heteroplasmy (proportion between normal and mutant mtDNA variants), mitotic segregation (mtDNA’s stochastic distribution during cell division), and the phenotypic threshold effect (tissue specific sensitivity to oxidative metabolism defects), are not sufficient to explain PMDs’ clinical heterogeneity [2,4,5]. For example, they cannot elucidate tissue specific alteration in individuals harboring homoplasmic mtDNA mutation, the absence of clear correlation between heteroplasmy level and disease severity, or the heterogeneous manifestations in nuclear DNA-related PMDs [4]. For mtDNA-related PMDs, environmental factors, other nuclear or mitochondrial mutations, mtDNA haplotype or tissue-specific nuclear gene expression patterns, and a complex molecular and metabolic cellular compensation response to OXPHOS defect may act as modifiers [2,4,5]. For nuclear DNA-related PMDs, the lack of complexity comprehension is even greater. It is important to underline that some of the abnormalities of nuclear genes involved affect mtDNA replication and the expression system (mtDNA maintenance), causing dysfunctions of the intergenomic signaling (manifesting with multiple mtDNA deletions or mtDNA depletion), and have combined features of mendelian and mitochondrial genetics [2]. Because of the vast clinical and genetic variability, diagnosing PMDs can be challenging. Given this, with this narrative review we aimed to dissect the most common “red flags” that could help clinicians in recognizing, diagnosing, and managing PMDs. For each red flag, we have outlined the pathogenetic mechanism, its presence in certain syndromic pictures, and the association with other phenotypic manifestations. For this purpose, to help the readers orient themselves in this maze, we summarized in Table 1 the main phenotypic characteristics of well-defined PMDs and their main genetic etiology.

2. Neurological Red Flags

2.1. Progressive External Ophthalmoplegia (PEO)

Chronic progressive external ophthalmoplegia PEO (cPEO) is a mitochondrial syndrome characterized by myopathy of extraocular muscles. Indeed, the extraocular muscles are dependent on oxidative metabolism and for this reason are highly vulnerable to mitochondrial dysfunction [4]. cPEO can initiate at any age with progressive bilateral eyelid ptosis and diffuse ophthalmoparesis. Typically, progressive ptosis represents the earliest and the most noticeable feature of PEO patients and leads to the gradual acquisition of a chin-up compensatory head position [6]. In addition, severe fatigue, depression, and pain might contribute to disease impact on the quality of life of PEO patients [7]. Moreover, dysphagia and decreased respiratory muscle strength, as well as obstructive apneas, can be present [8].
Indeed, the term “pure PEO” identifies patients with isolated ocular myopathy, while when PEO occurs with other symptoms of mitochondrial dysfunction with neuromuscular involvement, it is denoted as “PEO plus syndrome”. The symptoms more frequently associated with “PEO plus syndrome” comprise muscle weakness (43%), exercise intolerance (23%), muscle wasting (18%), hearing loss (15%), and swallowing impairment (15%). Furthermore, Orsucci and colleagues proposed the term “PEO-encephalomyopathy” to indicate patients with PEO combined with signs of central nervous system (CNS) pathology [9]. PEO can be even a clinical sign of a multisystem disease; indeed, it is one of the most frequent phenotypes of PMDs [9]. Particularly, PEO is a traditional clinical manifestation of Kearns–Sayre syndrome (KSS), together with pigmentary retinopathy and Pearson syndrome, together with sideroblastic anemia [10].
A retrospective study on a large cohort from the database of the “Nation-wide Italian Collaborative Network of Mitochondrial Diseases” showed that ocular myopathy was more often associated with mtDNA single large-scale deletions and POLG1 mutations, while it was less common in patients with OPA1 mutations and all the mtDNA point mutations, especially in tRNAs. Moreover, “PEO-encephalomyopathy” was often associated with the m.3243A>G mutation. Importantly, patients with POLG1 mutations seem to have more severe quality of life impairment compared to patients with mtDNA mutations or deletions [9].

2.2. Exercise Intolerance

Exercise intolerance is a feature which can alert clinicians to mitochondrial myopathies, though it is difficult to be clinically assessed. Indeed, it is an elusive symptom, which sometimes could be missed. However, exercise intolerance can be quantified through measurement of the maximal oxygen uptake, by evaluating resting and peak exercise-induced serum lactate levels, as well as by examining lactate/pyruvate ratios [11]. Owing to exercise performance limitations, patients with exercise intolerance can be mistaken for having cardiopulmonary diseases.
According to the Italian Network of Mitochondrial Disorders database, more than 20% of mitochondrial patients complain of exercise intolerance [12], which seems to be associated with specific mutations, as well as m.3243A>G. About one-third of patients with exercise intolerance symptoms showed increased creatine kinase (CK) levels. In addition, ragged red fibers (RRFs) and cytochrome c oxidase (COX)-negative fibers were often present in subjects with exercise intolerance [12].

2.3. Stroke-like Episodes (SLEs)

Stroke-like episodes (SLEs) are the defining features of mitochondrial encephalopathy with lactic acidosis and stroke-like episodes (MELAS) syndrome, a PMD often associated to multisystem involvement, such as diabetes, lactic acidosis, and hearing loss [10].
SLEs are characterized by headache, nausea and vomiting, encephalopathy, focal-onset seizures, or psychiatric disorders. In addition, epilepsia partialis continua and, sometimes, status epilepticus might even occur during the stroke-like episode. Due to its high energy demand, the brain is particularly vulnerable to mitochondrial oxidative phosphorylation defects [13]. A SLE, which can have an acute or subacute neurological manifestation, typically affects individuals younger than 40 years and it is caused by focal brain dysfunction of an epileptic nature. Late-onset SLEs have been rarely reported. [14].
About 80% of the cases are associated with the pathogenic m.3243A>G mutation in the MT-TL1 gene, the first genetic defect linked to MELAS syndrome [15]. However, other rare mtDNA mutations and recessive POLG1 mutations are well-recognized causes of SLEs [16,17]. Specifically, one-third of patients with the m.3243A>G mutation manifest SLEs after 40 year of age [18], whereas SLEs are more aggressive in patients harboring POLG1 mutations and appear earlier. Indeed, POLG1 patients have an explosive onset and a rapid progression, with a higher risk of death due to status epilepticus [17], in comparison to patients with 3243A>G mutation, who mainly die subsequently to complications not related to CNS. In particular, 3243A>G patients show a protracted disease course with an insidious onset [19,20]. Moreover, around half of POLG1 patients have no other symptoms outside of CNS features before the first SLE. On the contrary, m.3243A>G patients manifest preceding systemic features, which can be useful as “red flags” to forecast SLEs in PMDs associated with m.3243A>G mutation [20]. On brain magnetic resonance imaging (MRI) scans, SLEs show involvement of the cortex and juxtacortical white matter and typically are not confined to vascular territories. SLEs are characterized by an increased lactate level, both in the lesion area and in the apparently unaffected brain regions, evidenced by MRI proton spectroscopy, which can facilitate the screening and early diagnosis [3,21]. The neurological disability severity depends on the level of temporal, parietal, and/or occipital lobe involvement, evidenced through neuroimaging. Focal seizure activity can be detected by means of electroencephalography. Recurrent SLEs, usually followed by symptomatic complete or incomplete remissions, lead to progressive accumulation of neurological deficits.

2.4. Bilateral Brainstem Lesions

Bilateral brainstem lesions represent the typical neuropathological features of Leigh syndrome (LS), together with lesions of the cerebellum, diencephalon, and/or basal ganglia, which lead to a progressive decline in neurological function [10]. Indeed, bilateral symmetrical lesions extend from the basal ganglia and thalamus, through brainstem structures, to spinal cord posterior columns [22]. It should be noted that neurons become affected by an energy supply deficit because of their high energy demand [23].
LS is the most common presentation of pediatric mitochondrial disease and a progressive neurodegenerative disorder, affecting about 1 in 36,000 births [10,24]. Most affected patients die before 3 years of age, usually from sudden respiratory failure; indeed, the outcome of LS is poor [25]. The clinical hallmarks of LS include psychomotor delay or regression, hypotonia, tremor, weakness, truncal ataxia, and lactic acidosis [26], as well as dystonia [27]. There are even extra-CNS features in LS, as well as polyneuropathy, myopathy, diabetes mellitus, short stature, hypertrichosis, cardiomyopathy, anemia, sleep disturbances, renal failure, gastrointestinal dysfunction, hearing loss, failure to thrive, retinitis pigmentosa, cranial nerve palsies, and scoliosis. Leigh-like syndrome refers to patients with atypical symptoms, laboratory findings, and radiologic features, but with the overall clinical picture indicative of LS. In particular, Leigh-like patients manifest late-onset myopathy, and few or no ocular motility abnormalities, while a respiratory deficit is usually not present [28].
LS is an example of the genetic heterogeneity of PMDs. It is caused by mutations in about 100 genes across both nuclear and mitochondrial genomes [10]. Indeed, LS is commonly associated with mitochondrial complex IV (cytochrome c oxidase-COX) deficiency, which can result from mutations in several COX structural proteins or assembly factors, such as SURF1 [29], which represents the main cause of COX-deficient LS. Pathogenic variants in mtDNA-encoded complex I subunits or MT-ATP6 are strongly associated with subacute brainstem dysfunction in young patients [30,31]. Brainstem dysfunction has been evidenced even in patients with neuropathy and ataxia (NARP) associated with MT-ATP6 pathogenic variants, without a clear diagnosis of mitochondrial inherited LS (MILS) [30]. Noteworthy, La Morgia and colleagues reported an association between a rare variant (m.4171C>A) in MT-ND1, causing Leber’s hereditary optic neuropathy (LHON) and bilateral brainstem lesions together with vomiting and vertigo, in addition to the typical feature of LHON [32]. In addition, progressive brainstem lesions have been found even in patients with KSS [33,34].
At the microscopic level, neuronal loss, proportionate loss of myelin, reactive astrocytosis, and proliferation of cerebral microvessels are present [4]. A brain MRI with proton spectroscopy sequences is useful for diagnosis of LS. The affected regions appear symmetrical and hyperintense on T2-weighted sequences. Furthermore, increased lactate levels in the blood or cerebrospinal fluid represents another important clue for the diagnosis of LS [35].

2.5. Epilepsy

Epilepsy is one of the most common CNS features of PMDs. The pathophysiology of epilepsy in PMDs is not clearly understood, even if seizures can be explained mainly by an impaired oxidative phosphorylation leading to ATP deficiency [36].
Seizures are hallmarks in some PMDs, such as Alpers–Huttenlocher syndrome (AHS) [17], pyruvate dehydrogenase complex deficiency (PDHc) [37], LS [38], myoclonic epilepsy with RRFs (MERRF) [39], and MELAS syndrome [40]. The incidence rate of epileptic seizures among adult PMDs patients is about 10–40% and is 60% in pediatric patients [41]. PMD patients often experience myoclonus and various types of focal seizures, or even tonic seizures, tonic–clonic seizures, and infantile spasms. Moreover, patients manifest status epilepticus, including nonconvulsive status epilepticus and epilepsia partialis continua, which is a focal motor status epilepticus occurring for a minimum of one hour and recurring at ten seconds intervals [36,42]. Interestingly, status epilepticus is more common in mitochondrial depletion syndromes (MDS) or mtDNA-related PMDs, and usually coexists with SLEs [36]. Of note, epilepsy in MELAS is heterogeneous; however, focal onset, clonic, and tonic seizures represent the most common SLEs-associated epileptic phenomena [43].
It is noteworthy that repeated or long-lasting seizures prolong the energy deficit, leading to further mitochondrial dysfunction, which conducts to subsequent seizures. Indeed, this self-perpetuating cycle may lead to the development of SLEs, especially in MELAS and POLG-related diseases, often related to status epilepticus.
Mitochondrial epilepsy is usually challenging to treat, often leading to progressive epileptic encephalopathy and a worse prognosis [36]. In particular, epilepsy due to the mutation in the POLG1 gene is refractory to pharmacotherapy [44]. Regarding treatments, epilepsy associated with PMDs can be managed as non-mitochondrial epilepsy [43]. Indeed, levetiracetam in association with benzodiazepines is the first line therapy for myoclonus in MERFF [36], whereas lamotrigine might promote myoclonic seizures [45]. Patients with MELAS or drug-resistant seizures can be treated with zonisamide and lacosamide [36,43]. Additionally, midazolam, along with alternative interventions, as well as perampanel, corticosteroids, ketamine, immunoglobulin, and magnesium infusion are employed for the treatment of status epilepticus [43,46,47]. To obtain seizure control, most patients need to be treated with multiple anti-epileptic drugs and therapy must be personalized for each patient. Nevertheless, it is important to not use drugs with mitochondrial toxicity, such as sodium valproate, especially in patients harboring POLG1 mutations in order, to avoid seizure aggravation and hepatic failure [48]. Currently, new therapies focused on antioxidant approaches, mitophagy and mitochondrial biogenesis, or stabilization of the mitochondrial membrane are under investigation [49].
Importantly, there is no typical electroencephalography trace for seizures in PMDs, even if background activity is usually disturbed. Regarding neuroimaging, the analysis of 1467 patients with PMDs revealed that MRI abnormalities were significantly more common in subjects with epilepsy compared to patients without seizures [50]. Indeed, brain atrophy, stroke-like lesions, basal ganglia, and white matter changes are more common in PMDs patients with epilepsy [51].

2.6. Movement Disorders

Movement disorders in PMDs are not rare and, even when isolated, these should incite a search for PMD diagnosis. In particular, ataxia and parkinsonism represent the most common movement disorders [52]. Muscle and nerve cells have high energy needs and ATP derived from mitochondria represents their main source of energy [53]. Indeed, neurological and muscular symptoms are common features of PMDs. Cerebellar ataxia, one of the most prevalent neurological mitochondrial signs [52], is characterized by movement incoordination or imbalance and leads to abnormal movement and gait, caused by dysfunction of the cerebellum or its connections [54]. Cerebellar ataxia is often associated with abnormal eye movements, dysmetria, kinetic tremor, dysarthria, and/or dysphagia, other symptoms of cerebellum dysfunction [10]. However, sensory ataxia is characterized by an alteration in the proprioceptive system due to spinal or peripheral lesions. Stepping gait, which aggravates with loss of visual fixation, is a feature of sensory ataxia [52]. Cerebellar ataxia can be the onset symptom of well-defined mitochondrial syndromes, such as KSS, MILS, MERRF, and can also be one of the clinical symptoms in undefined mitochondrial encephalopathies [55]. In addition, isolated ataxia may even be due to autosomal recessive gene defects, including POLG-related diseases, such as mitochondrial recessive ataxia syndrome (MIRAS) [56]. Autosomal recessive coenzyme Q10 deficiency, a treatable PMD, is characterized by isolated cerebellar ataxia. In addition, ataxia can be associated with ocular myopathy and with psychiatric comorbidities or, rarely, epilepsy [57], such as in infantile-onset spinocerebellar ataxia (IOSCA), a neurodegenerative disorder caused by TWNK mutations [10,58]. Sensory ataxia is a less common feature among PMDs and mainly characterizes neuropathy, ataxia, and pigmentary retinopathy syndrome (NARP) and sensory ataxia neuropathy dysarthria and ophthalmoplegia (SANDO) [10,59]. Recently, recessive mutations in the PTRM1 gene, encoding for a mitochondrial matrix enzyme involved in mitochondrial proteostasis, were found to be associated with complex early-onset phenotypes including spinocerebellar ataxia [60]. Neuroimaging may be supportive for the diagnosis; indeed, a brain MRI can show peculiar alterations depending on the syndrome, together with cerebellar atrophy [52].
Mitochondrial parkinsonism is usually difficult to diagnose due to similarities with idiopathic Parkinson’s disease. Usually, the disease onset is at around fifty years of age [10]. Both nuclear gene mutations and mtDNA genetic abnormalities can cause mitochondrial parkinsonism. Between nuclear genes, the most prevalent are POLG1 and TWNK [54]. These genes should be tested, especially in families with autosomal dominant PEO and parkinsonism. Levodopa-responsive parkinsonism has been reported in patients with either biallelic or heterozygous POLG1 mutations [61]. Among mtDNA genetic abnormalities, the m.8344A>G mutation in the MT-TK gene, causing MERRF syndrome, was described in one patient with levodopa-responsive parkinsonism [62]. Furthermore, MT-ND1, MT-ND6 gene mutations and a 4-base-pair deletion in the cytochrome B gene were found in patients with parkinsonism associated with optic atrophy, myoclonic epilepsy and SLEs, typical PMDs red flags [63,64,65]. To conclude, the diagnosis of mitochondrial parkinsonism should have to be accounted for in the presence of other signs of mitochondrial dysfunction, as well as PEO, neuropathy, myopathy, ataxia, isolated muscle pain, psychiatric disorders, and epilepsy.
Noteworthily, along with ataxia and parkinsonism, in a large cohort study, Montano and colleagues reported other movement disorders, such as tremor and dystonia, as clinical features of adult-onset PMDs [66].

2.7. Migraine

Migraine represents another significant neurological red flag of PMDs [10]. Mitochondrial dysfunction plays a central role in the pathophysiology of migraine, which reflects the vulnerability of CNS to respiratory chain defects. However, clear pathomechanisms are not delineated [67]. Importantly, due to the absence of a structured diagnostic headache tool, specific questionnaires, interviews, and objective clinical examination, the prevalence of migraine in PMDs patients fluctuates noticeably in the scientific literature [68]. A cross-sectional cohort study reported that migraine has a prevalence of 35% in PMDs patients, which is higher compared to the general population. This prevalence was reported to be independent from sex, phenotype, or genotype [68]. Moreover, the same study observed that epilepsy, myoclonus, and SLEs are more frequent in patients with migraine compared to patients without it. Noticeably, migraine might be an early manifestation of the disease [68]. Specifically, MELAS patients harboring m.3243A> G point mutation can suffer from repeated episodes of migraine, which is one of the most frequent symptoms [69,70]. Noteworthily, pharmacological management of PMDs patients with migraine could be particularly challenging, probably due to poor tolerance to drugs commonly used for headaches [71].

2.8. Neuropathy

Peripheral neuropathies occur in a third of patients with PMDs. Peripheral nerves are highly dependent on energy metabolism, as well as mitochondria that play a pivotal role in maintaining viability of motor and sensory neurons and their axons [72]. Neuropathies are particularly associated with defects in mitochondrial DNA maintenance and replication, as well as defects in the respiratory chain, such as complex V. However, the number of genes and phenotypes linked with PMDs and neuropathies is rapidly increasing [72].
Peripheral neuropathy can manifest as a minor clinical sign in the majority of PMDs, with a small impact on patients’ lives. However, moderate to severe peripheral neuropathy might be the first manifestation in specific syndromes, such as KSS, LHON plus, mitochondrial neurogastrointestinal encephalomyopathy (MNGIE), MERRF, MELAS, and LS [72,73,74,75,76,77,78]. Peripheral neuropathy associated with PMDs mainly manifests as chronic sensorimotor axonal polyneuropathy [79]. Specifically, sensory ataxic neuropathy, presenting in isolation or in combination with other signs/symptoms, is usually associated with NARP and POLG-related disorders (especially SANDO phenotype).
Mutations in the MFN2 and GDAP1 genes encoding proteins involved in mitochondrial dynamics have been evidenced as causes of Charcot–Marie–Tooth neuropathy (CMT) [80]. CMT neuropathy represents the most common inherited neurological disorder, affecting around 1 in every 2500 people in the world [81,82]. The typical CMT phenotype comprises distal weakness, sensory loss, foot deformities, as well as absent ankle reflex, manifesting in most patients in the first or second decade of life. However, many patients develop severe disability in infancy or early childhood and there are patients which manifest few or no symptoms of neuropathy until adulthood [82]. The majority of CMT neuropathies are demyelinating, while up to one-third are the result of a primary axonal disorder [83,84]. Mutations in MFN2 and GDAP1 affect axonal mitochondrial transport and function and have been associated with axonal forms of CMT. The mitochondrial dynamics alterations affect peripheral axons from spinal cord motor and dorsal root ganglia neurons, causing peripheral neuropathies [85].
Noteworthily, muscle biopsy, neuroimaging, cerebrospinal fluid analysis, and metabolic and genetic tests in blood and muscle are essential for the diagnosis of peripheral neuropathies [82].

2.9. Cognitive Decline

Cognitive decline may represent a clinical manifestation of PMDs, even if it remains one of the least understood aspects [86]. How the cognitive state might change with PMDs’ progression remains unclear. Most of the studies reporting cognitive impairment in PMDs patients are mainly retrospective analyses of patients with high genotypic variability and additional risk factors [87]. According to El-Hattab and colleagues < 10% of MELAS patients showed developmental delay as am initial clinical manifestation, while 10–24% of MELAS patients manifested impaired mentation during the disease course. In addition, 50–74% of MELAS patients revealed learning disability or memory impairment, while ≥90% of patients experienced dementia [40]. However, generalizing these findings to a wider population of PMDs patients is challenging [88,89]. More recently, disease severity has been demonstrated to be a stronger predictor of cognitive abilities compared to genotype [87]. Importantly, Kaufmann and colleagues [90] found a progression of general cognitive difficulties over a 4-year period in fully symptomatic MELAS patients with the m.3243A>G genotype. In addition to MELAS, cognitive impairment may be a clinical feature of other PMDs, such as MERRF syndrome [91], LS [92], NARP [93], and LHON [94]. Cognitive decline is also a prominent feature in PTRM1-related phenotypes. Interestingly, PITRM1 mutations have been found to cause cerebral amyloid beta accumulation, significantly linking mitochondrial functioning with amyloidotic neurodegeneration [60]. Noteworthily, brain MRI findings varied considerably between those studies, leading to a weak association between cognitive impairments and neuroimaging results [87].

2.10. Optic Atrophy and Pigmented Retinopathy

Common clinical features of PMDs include ocular manifestations, such as optic atrophy and retinopathy. Mitochondria are essential for the survival and function of retinal ganglion cells (RGCs), one of the neuronal populations of the retina. Given their strong dependency on oxidative metabolism, RGCs are particularly susceptible to mitochondrial deficit [95].
LHON is the most common inherited optic nerve disease, hallmarked by subacute bilateral loss of central vision due to dysfunction and loss of RGCs [96]. LHON primarily affects healthy young males compared to females [97]. Indeed, gender is a risk factor, partially explainable by the protective effect of estrogens [98]. Environmental factor, such as smoking, play a role in LHON pathogenesis [99]. The onset of visual loss in LHON is usually unilateral, with the second eye becoming affected within weeks or months [3]. Optic disc hyperemia, oedema of the peripapillary retinal nerve fiber layer, and retinal telangiectasia represent typical ophthalmological features in the acute phase of LHON [100]. Furthermore, thickening of the retinal nerve fiber layer has been described before angiopathic changes [101] LHON is caused by mtDNA point mutations, with the m.11778G>A in MT-ND4 being the most common and the one associated with poorer outcomes [102]. Other common LHON mutations associated with LHON disease include m.3460G>A in MT-ND1 and m.14484T>C in MT-ND6 mutations [103,104]. Thus, the vast majority of LHON are accounted by three mtDNA mutations encoding for mitochondrial complex I subunits.
Autosomal dominant optic atrophy (ADOA) is another PMD affecting RGCs. As with LHON, ADOA is characterized by central vision deterioration caused by optic nerve atrophy and retinal nerve fiber layer degeneration [105]. ADOA is associated with mutations in the nuclear-encoded gene OPA1, which is involved in several cellular processes, including the mitochondrial dynamics [10]. Indeed, around 50–60% of ADOA cases are caused by OPA1 mutations, with a prevalence of 1 in 25,000 people [100]. Blindness occurs in 85% of ADOA patients, manifesting at a mean age of 8 years [10]. As with LHON, in about 20% of cases, ADOA patients can manifest extra-ocular neurological phenotypes (ADOA-plus) [4]. The most frequent “plus” manifestation is bilateral sensorineural deafness, described in late childhood and early adulthood; other clinical manifestations, such as ataxia, peripheral neuropathy, and ocular and skeletal myopathy might appear later, in the third decade of life. These patients’ muscle biopsies show multiple mtDNA deletions and COX-deficient fibers, given the OPA1 role in mt-DNA maintenance [106]. The proportion of COX-negative fibers is much higher in patients with ADOA-plus compared to non-syndromic ADOA patients [4].
Pigmentary retinopathy, resulting from retinal degeneration, may occur in PMDs patients. It is characterized by progressive photoreceptor damage and subsequent cell death, leading to the atrophy of photoreceptors and adjacent retina layers [107,108]. mtDNA single large-scale deletions are associated with diffuse pigmentary retinopathy [109]. Indeed, pigmentary retinopathy, together with PEO, is a primary clinical feature of KSS [10], whereas mtDNA point mutations are mainly related to clinically more localized macular pigmentary and atrophic changes [107]. Therefore, patients harboring m.3243A>G mutation may manifest pigmentary retinopathy as the most common ocular sign [110]. During ophthalmoscopy investigation, pigmentary retinopathy is defined by the presence of retinal pigment epithelium, hyperpigmented areas close to the back of the eye. However, pigmentary retinopathy can be even asymptomatic, remaining undiagnosed. Finally, pigmentary retinopathy can also occur in patients with LS or cPEO, as well as in patients with maternally inherited diabetes and deafness syndrome (MIDD) [111,112,113].

2.11. Sensorineural Hearing Loss

Hearing impairment is a common clinical feature of PMDs, both as an isolated sign and as part of a multisystem phenotype. Usually, hearing loss in PMDs manifests at a young age after speech acquisition (post-lingual) with other systemic clinical signs of PMDs, as well as with a maternal pattern of inheritance [114,115]. Sensorineural hearing loss is mainly caused by degeneration and loss of hair cells in the cochlea of the inner ear, which are highly dependent on mitochondrial metabolism [116,117].
The mitochondrial non-syndromic hearing loss (NSHL) is an example of isolated manifestation of hearing loss, which is associated with several mtDNA variants, especially in MT-RNR1 or MT-TS1 [118]. Moreover, sensorineural hearing loss can be part of a mitochondrial syndrome spectrum, such as MELAS, KSS, MERFF, and MIDD [10]. Indeed, sensorineural hearing loss may be a clinical feature in patients with POLG1 and SPG7 mutations [114]. In addition, hearing loss represents the most prevalent extra-ocular clinical feature in ADOA associated with the OPA1 gene [115].
Due to their prokaryotic origins, mitochondria are vulnerable to antibiotics which target the bacterial ribosome, particularly in presence of specific mtDNA mutations. Specifically, patients harboring m.1555A>G or m.1494C>T in MT-RNR1 may manifest deafness after exposure to aminoglycoside antibiotics. Indeed, the screening test for mtDNA mutations could be useful before starting the treatment [119].

3. Gastrointestinal Red Flags

3.1. Liver Failure

Acute or fulminant liver failure is a significant indicator of PMDs. In particular, mitochondrial liver disease manifests primarily in children, either in isolation or association with CNS involvement [120]. Mitochondria represent the major energy source for liver cells; indeed, impaired mitochondrial functions may contribute to liver damage [121]. Recessive mutations in the nuclear-encoded gene TRMU, encoding for a tRNA-modifying protein, may lead to acute liver failure in the first months of life [122].
Liver disease associated with chronic neuromuscular disease or other multisystem involvement might be a clinical sign of PMDs. Liver failure is a clinical feature of AHS, a severe pediatric encephalopathy, associated with recessive POLG1 mutations [123]. AHS is hallmarked by liver degeneration and encephalopathy characterized by intractable seizures, with frequent episodes of epilepsia partialis continua or status epilepticus, and by progressive developmental regression. Liver failure may precede or occur after seizure onset. Terminal liver impairment is common [124]. Sodium valproate treatment should be avoided in AHS because it might precipitate liver failure [119,125]. Other MDS, such as recessive TWNK mutations, can be associated with signs of liver impairment, together with severe early onset encephalopathy, describing a phenotype reminiscent of AHS. In particular, clinical features include hypotonia, athetosis, sensory neuropathy, ataxia, hearing deficit, ophthalmoplegia, intractable epilepsy, and elevation of serum transaminases. Liver biopsy shows mtDNA depletion, whereas the muscle mtDNA can be only slightly affected [126]. Mutations in DGUOK and MPV17, two genes encoding mitochondrial proteins involved in mtDNA maintenance, cause infantile hepatocerebral MDSs [127]. Mutations in DGUOK, encoding for the mitochondrial deoxyguanosine kinase, specifically causes MDS type 3, while MPV17 is associated with MDS type 6 [128]. MDS type 3 is characterized by early-onset severe liver disease, low-birth weight, and neurological impairment, such as myopathy and nystagmus, whereas MDS type 6 is characterized by infantile or childhood onset progressive hepatic disease and systemic manifestations, such as hypoglycemia and lactic acidosis. In both cases, liver dysfunction progress to chronic liver failure, the most common cause of death in these PMDs [129]. According to some investigations the levels of tyrosine and phenylamine are elevated in the blood or urine of newborns harboring DGUOK and MPV17 mutations [130,131]. Mutations in ETHE1 can also cause liver failure. ETHE1 encodes for a mitochondrial protein that plays a key role in hydrogen sulfide detoxification as a sulfur dioxygenase. ETHE1 mutations lead to the toxic accumulation of H2S and metabolites in tissues and body fluids, causing a severe fatal metabolic disorder known as ethylmalonic encephalopathy, a complex disorder characterized by, in addition to liver dysfunction, chronic diarrhea, petechial rush, and neurological manifestations [132,133]. Liver transplantation could increase the survival of patients with DGUOK and ETHE1 mutations [134,135].

3.2. Severe Dysmotility/Pseudo-Obstructive Episodes

Gastrointestinal dysmotility, due to degeneration of gastrointestinal tract muscles, is a red flag for PMDs, usually encountered with multiple genetic defects [3]. The pathophysiology underling this phenomenon remains unclear [136]. Progressive gastrointestinal dysmotility is a severe clinical feature of MNGIE, in combination with neuromuscular involvement (PEO, polyneuropathy, hearing loss, and leukoencephalopathy) [137]. Gastrointestinal signs/symptoms comprise diarrhea, abdominal pain, vomiting, pseudo-obstruction which may lead to weight loss, and cachexia.
MNGIE is an autosomal recessive syndrome caused by mutations in the TYMP gene leading to thymidine phosphorylase deficiency. It usually manifests at a mean age of 18 years and leads to death in early adulthood. MNGIE is frequently misdiagnosed; according to Hirano and colleagues, there is limited evidence regarding diagnostic and therapeutic approaches due to the lack of controlled studies with proper follow-up [138]. Interestingly, an observational cohort study reported that severe gastrointestinal pseudo-obstruction is much more common in PMDs patients with the pathogenic m.3243A>G mtDNA mutation, despite the fact gastrointestinal symptoms are usually under-recognized in PMDs. For example, pseudo-obstructive episodes are common in MELAS patients, especially during the acute phase of a SLE. Considering the frequency of m.3243A>G mtDNA mutation, clinicians should always assess the gastrointestinal system in those patients to mitigate the risk of developing severe intestinal pseudo-obstructions [139].
A clinical phenotype close to MNGIE syndrome has been reported in five patients with POLG1 mutations, who had prominent symptoms of persistent diarrhea and cachexia related to gastrointestinal dysmotility, as well as ptosis, proximal myopathy, and sensory neuropathy [140,141,142]. Finally, gastrointestinal dysmotility, feeding difficulties, and failure to thrive are gastrointestinal manifestations observed in the infantile hepatocerebral MDSs due to MPV17 mutations [143].

4. Cardiovascular Red Flags

4.1. Cardiomyopathies

The heart is one of the most metabolically active tissues in the human body, relying heavily on mitochondrial ATP production to maintain the high energy demand of individually contracting cardiac myocytes. Adults with PMDs are indeed about 30% more likely to have abnormal electrocardiograms and/or echocardiograms.
During cardiac MRI, more than one-third of these patients exhibit late gadolinium enhancement linked to myocardial fibrosis. Cardiomyopathies are the most frequent cardiac manifestations of PMDs and are estimated to occur in 20–40% of patients [144]. Hypertrophic cardiomyopathies are most common, but dilated, restrictive, and other types are also seen. The severity ranges from asymptomatic, sometimes spontaneously reversible conditions to a severe cardiomyopathy with an early, even prenatal, onset that causes death in early infancy. The presence of cardiomyopathy in a PMD, regardless of its severity, is associated with a poorer prognosis [145]. Cardiomyopathies have been reported in patients with MELAS in 30–32% of cases [70] and LS in 18–21% of cases. In LS patients with cardiac manifestations, hypertrophic cardiomyopathy was found in 50% of cases [146].
Two syndromic cardiomyopathies presenting in infancy are Barth syndrome and Sengers syndrome. Barth syndrome is an X-linked condition characterized by dilated cardiomyopathy, skeletal myopathy (mostly proximal), poor growth, (cyclical) neutropenia, and 3-methylglutaconic aciduria [147]. Hypertrophic cardiomyopathy, left ventricular noncompaction, and endocardial fibroelastosis are further cardiac characteristics. Barth syndrome is caused by mutations in the TAZ gene, which alter the inner mitochondrial membrane’s ability to produce cardiolipin [148].
Mutations in the gene encoding for acylglycerol kinase (AGK), which is also a part of the mitochondrial import machinery, cause Sengers syndrome, a rare autosomal recessive PMD [149,150]. Hypertrophic cardiomyopathy and congenital cataract are present in affected newborns. Lactic acidosis, exercise intolerance, and skeletal myopathy are additional clinical characteristics. Infant mortality from progressive heart failure is possible, but long-term survival has been observed in some patients.
Additionally, other causes of childhood onset mitochondrial cardiomyopathies are increasingly recognized, including disorders of mitochondrial translation and defects of coenzyme Q10 biosynthesis [151].

4.2. Arrythmias, Cardiac Conduction Defects, and Valvulopathies

Arrythmias, conduction defects, and pulmonary hypertension are examples of other rarer cardiac manifestations [152]. Prolonged QT and arrhythmias are common in patients with cPEO [153]. Cardiac conduction defects, often culminating in a complete heart block, have been reported frequently in patients with KSS. Biallelic variants in PPA2 gene, encoding for a mitochondrial located pyrophosphatase, have been associated with sudden death [154]. According to a recent systematic review, patients with MELAS have significantly more electrocardiography and echocardiography alterations compared to patients with other PMD phenotypes [155]. A well-described cardiac complication in MELAS patients is represented by Wolff–Parkinson–White syndrome [155] a conduction abnormality characterized by the presence of altered electrically conductive circuits between the atria and ventricles. Cardiac conduction defects may also be present in MERRF, which is caused by m.8344A>G mutations in MT-TK in 80–90% of cases [156,157]. One case series found cardiac abnormalities in 53% of MERRF patients harboring this mutation [158]. The m.8363G>A mutation has also been reported in a case series of nine patients with clinic presentations fitting MERRF, and four of the nine patients were found to have cardiac conduction abnormalities [159].
Despite being a rare manifestation of PMDs, valvular heart disease has been linked to PDSS1 mutations, which encodes for an enzyme that produces coenzyme Q10 [160]. Recently, it has been suggested that children with LS elicited by ADAR mutations are susceptible to developing fatal heart valve calcification [161].

5. Kidney Red Flags

5.1. Tubulopathies

The kidney primarily depends on aerobic metabolism. Due to its inability to synthesize ATP anaerobically, the cortical tubule, especially in its proximal part, is extremely susceptible to OXPHOS [162]. Numerous nuclear and mtDNA gene mutations have been linked to renal manifestations in PMDs. The most frequent condition is tubular dysfunction, which can range from a mild hyperaminoaciduria triggered by catabolic circumstances (i.e., sepsis, fever) to a full-blown de Toni–Debré–Fanconi syndrome. Depending on the location of tubule damage and the type of chemicals lost in the urine, the tubulopathy may have Fanconi- or Gitelman-like characteristics. Gitelman syndrome affects the distal tubule and causes more specialized losses, whereas Fanconi syndrome affects the proximal tubule and causes broader losses (i.e., glucosuria, phosphaturia, generalized aminoaciduria, and bicarbonate wasting). Both diseases can produce hypomagnesemia, but Gitelman syndrome is more commonly associated with it [163]. The most severe tubulopathies are typically associated with large-scale mtDNA deletions and KSS or Person syndrome phenotypes [164]. Moreover, MDSs (especially RRM2B defect but also DGUOK, MPV17, SUCLA2, and TK2 mutations) and disorders of mitochondrial translation (such as TFSM gene mutations), are also associated with tubulopathy [165].

5.2. Glomerulopathies

Focal segmental glomerulosclerosis and steroid-resistant nephrotic syndrome have been reported in PMDs patients with the m.3243G>A mutation [166,167]. Focal segmental glomerulosclerosis is frequently observed in adults but rarely in children [167]. A pseudohypoaldosteronism-like picture with hyperkaliemia, hyponatremia, and gradual renal impairment can occur in children with RMND1 mutations [168]. Some defects in the CoQ biosynthesis pathway (PDSS2, COQ2, COQ6, COQ8 deficiencies) are also associated with glomerular disease and may respond to treatment with CoQ10 [169].

6. Endocrine Red Flags

6.1. Diabetes Mellitus

Because steroid hormones are synthesized in the mitochondria, reduced ATP production affects hormone production and results in endocrinological symptoms. Overall, it appears that abnormalities brought on by mtDNA flaws, particularly large-scale deletions and point mutations in tRNA genes, are more likely to have endocrinological symptoms. These symptoms may also be present in patients with nuclear gene abnormalities, which most typically involve mtDNA maintenance and translation-related problems [170]. The endocrine manifestation that is best characterized is diabetes mellitus. The impairment of the mitochondria’s function as a glucose sensor, which links glucose metabolism to insulin release, contributes to the mechanism of diabetes in PMDs in addition to decreased insulin secretion brought on by a lack of ATP supply [171]. Diabetes is reported in a substantial proportion of patients carrying the mutation m.3243A>G in MT-TL1, either as a dominant feature in MIDD, in multisystem phenotypes, or as a part of MELAS. The m.3243A>G mutation is estimated to cause 0.5–2.9% of diabetes mellitus in the population [170]. Diabetes mellitus is also frequently seen in KSS or in Pearson syndrome, where exocrine pancreas dysfunction is a more prominent feature [172].

6.2. Other Endocrine Signs

Additional endocrinological manifestations that should be mentioned are hypothyroidism, hypoparathyroidism, adrenal insufficiency, and hypogonadism [170,173,174]. Premature ovarian failure is observed in POLG-related diseases and in association with leukoencephalopathy in AARS2 deficiency. Although isolated endocrine involvement may be the first sign of a mitochondrial problem, mitochondrial endocrine dysfunction most usually arises in the context of multisystem disease [170].

7. Other Red Flags

7.1. Lactic Acidosis

Lactic acidosis is the most well-known laboratory finding in patients with PMDs. ATP synthesis is reduced because of malfunction in the electron transport chain. Low ATP levels activate glycolysis, which overproduces pyruvate. Extra pyruvate is either converted to lactate or transaminated to alanine. Congenital lactic acidosis has most frequently been reported in children with genetically determined complex I deficiency [175]. However, lactic acidosis has historically being recognized as a feature of different PMDs. Lactic acidosis may be transient and resolve within a few days of birth in some PMDs [176]. A total of 50% of 51 PMDs patients with different ages investigated by Jackson and colleagues had lactic acidosis [177]. Patients with mtDNA abnormalities are more likely to develop lactic acidosis [178], and mtDNA mutations are responsible for 10% of all PMDs in children [179]. In a study performed by Hutchesson and colleagues in 11 infants with known PMD, increased plasma levels of lactate were found [180]. Munnich and colleagues [178] carried out the biggest investigation of lactic acidosis, specifically in childhood-onset PMDs. A higher venous lactate level was detected in 30% of the 235 children investigated.
Although common in children, severe lactic acidosis is rare in adults with PMDs. In adults, lactic acidosis is frequently observed associated with other mitochondrial manifestations, such as stroke-like episodes, encephalopathy, seizures, and myopathies [181]. Lactic acidosis is a low-specificity biomarker: it can be caused by, among others, venous stasis, hypoxia, hypoperfusion, hepatic dysfunction, renal failure, drug toxicity, sepsis, spasticity, hyperinsulinism, chronic thiamine deficiency, and seizures [178,180].

7.2. Cachexia

Cachexia is a wasting illness elicited by mitochondrial disfunctions, so occurs in PMDs, especially associated with MELAS or MNGIE [137]. Cachexia is a syndrome that, by definition, affects skeletal muscle mass and function with or without body fat loss [182]. Furthermore, cardiac dysfunction brought on by striated muscle atrophy, as well as respiratory distress due to its effects on respiratory muscle function, are both primary causes of death [4]. Numerous inflammatory cytokines are involved in the etiology of cachexia, including tumor necrosis factor alpha, interleukins 1 and 6, and interferon gamma [182]. Systemic inflammation is one of the major underlying mechanisms driving the reduction in skeletal muscle mass [183]. In rodents, early impairment of mitochondria quality control and function has been reported [183,184] and has been observed to trigger events occurring before muscle atrophy in cachexia-related muscle atrophy [185]. In animal models of cachexia, the enhanced activity of the ubiquitin–proteasome pathway may drive muscle wasting [183,186]. In humans, activation of this system is not a consistent finding, suggesting the activation of other pathways, such as autophagy [187] and apoptosis [188].

7.3. Lipomas

Rarely, PMDs may present with multiple systemic lipomatosis (MSL) a rare disorder involving the adipose tissues and characterized by the development of non-encapsulated lipomas usually distributed in the cervical, cranial, and thoracic region. MSL has been reported in patients harboring point mutation in the mtDNA-encoded tRNA-lysine gene (MT-TK), which is the most frequent mutation associated with MERRF [189]. Muscle biopsies from roughly 28% of MSL patients revealed mitochondrial impairments, such as RRFs and COX-negative fibers [190]. The m.8344 A>G mutation in MT-TK has been identified in about 16% of MSL patients [191,192]. Additionally, MSL has recently been identified in a family with mtDNA multiple deletions harboring a mutation in the MFN2 gene [193]. Furthermore, MSL was associated with PEO and cerebellar ataxia, but not with diabetes or dyslipidemia, in a cohort of Italian PMDs patients. Additionally, mitochondrial MSL is more common in women and those who have never abused alcohol, is frequently linked to muscle involvement, and can be asymmetrical. A mitochondrial dysfunction mechanism is clearly supported by the correlation between MSL and high lactate serum level.

7.4. Short Stature

A major physiological endpoint is the growth and multiplication of chondrocytes in the growth plate, which is under the influence of many stimuli during childhood and adolescence. The development process may be compromised by mitochondrial malfunction. Children with PMDs are roughly shorter than their counterparts who are not afflicted, according to Wolny and colleagues [194], and their body mass index (BMI) is also reduced as a result [195]. In a Chinese cohort of pediatric patients with the m.3243A>G mutation in MT-TL1, 73% of the children were found to have short stature [196]. Short stature and poor growth are common in different PMDs, especially MERRF, MELAS [10], NARP [197], Pearson syndrome, and KSS [10]. Intriguingly, patients with short stature are more likely to have diabetes and cardiovascular involvement, whereas patients with low BMI are associated with neurological symptoms, such as seizures, encephalopathy, and SLEs [139]. Short stature and low BMI are also linked to gastrointestinal problems, as well as hearing loss.
Moreover, numerous cases of growth-hormone insufficiency in patients with PMDs caused by m.3243A>G, single large-scale mtDNA deletions, and nuclear-encoded gene mutations have been reported [198,199]. One of the proposed theories is that mitochondrial alterations can affect pituitary function [200]. However, other factors may contribute to short stature and low BMI, such as lower oral intake due to poor appetite and early satiety linked to gastrointestinal dysmotility, gastroparesis, delayed stomach emptying, bacterial overgrowth, intestinal pseudo-obstruction, and dysphagia [201]. PMDs patients may develop malabsorption, and mtDNA abnormalities have been demonstrated to disrupt the function of the human colonic epithelium [202].
Patients with PMDs could also experience additional comorbid conditions, which could hypothetically have an impact on bone health and growth [170,203]. Endocrinopathies, including hypoparathyroidism, hypothyroidism, and diabetes, are among them. Growing evidence suggests that growth may be impacted by systemic inflammation and stress in chronic illness states [204]. Hypothyroidism is rare in adults with PMDs, and mitochondrial diabetes typically appears beyond the age of 35, well past the period of childhood and adolescence when growth occurs [205].

8. Combinations of Symptoms

Certain constellations of symptoms may also be clues to specific PMDs. For example, the combination of ataxia and myoclonus, traditionally linked to the progressive myoclonus–ataxia syndrome and progressive myoclonus epilepsy spectrum, may represent a red flag for MERRF syndrome. The combination of ataxia and myoclonus with other manifestations, such as SLEs, hearing loss, diabetes, optic atrophy, and cognitive impairments, may be related to MELAS [10]. Another example is parkinsonism, which may suggest a mitochondrial etiology in the case of association with optic atrophy or PEO [206]. The combination of diabetes mellitus and sensorineural hearing loss can be the manifestation of different PMDs, especially if diagnosed at young age in patients with low or normal BMI [207]. For example, the maternal heritability of diabetes or impaired glucose tolerance in addition to hearing impairment and maculopathy may be a red flag for MIDD, which is typically caused by m.3243A>G [208]. However, the association of diabetes and deafness is also observed in other non-mitochondrial syndromes, such as Wolfram syndrome (diabetes insipidus, diabetes mellitus, optic atrophy, and deafness (DIDMOAD)) [209], Rogers syndrome (megaloblastic anemia, diabetes mellitus, and sensorineural deafness) [210], or Herrmann syndrome (photomyoclonus, diabetes mellitus, deafness, and nephropathy) [211].
Finally, the combination of hepatopathy and encephalopathy occurring in patients with acute or chronic liver failure is related to MDSs. Affected infants typically present with growth failure, feeding difficulty, developmental delay, and hypotonia. Although brain involvement is usually prominent, in rare cases, DGUOK mutations may cause isolated liver failure. In POLG-related diseases with hepatopathy, brain involvement may not be immediately apparent, but developmental regression due to epileptic encephalopathy is inevitable [212].

9. Focus on Pediatric Mitochondrial Neurological Red Flags

The neurological features or symptoms present in the majority of children affected by PMDs are muscle weakness, peripheral neuropathy, ophthalmoplegia, movement disorder, epilepsy, and migraine, which illustrate why the neuropediatrician is one of the key physicians involved in diagnosis and management of childhood-onset PMDs.
Seizures are a frequent complication of pediatric PMDs. PMDs patients may also present with epileptic syndromes, such as West and Lennox–Gastaut syndrome [213]. AHS, due to recessive mutations in POLG1, is the most common pediatric PMDs associated with epilepsy [214]. AHS patients often show focal, myoclonic, or complex seizures. Status epilepticus is common, sometimes starting with epilepsia partialis continua, followed by a generalized, therapy-resistant status.
Gross and/or fine motor delay are also frequent complications in pediatric patients and may be linked to hypotonia or myopathy. Myopathy is often part of encephalomyopathy but can also occur as a pure manifestation. Usually, the term “floppy infant” is used to describe a newborn with poor muscle tone and weakness [215].
In the pediatric population, among movement disorders, dystonias are the most frequent symptoms, and are particularly seen in LS [27]. This is not surprising, as the syndrome includes lesions in the basal ganglia and other extrapyramidal structures, from which these types of symptoms arise.
Another nonspecific CNS symptoms is developmental delay [216], which is usually global and affects cognitive, language, and motor skills. Given the enormous heterogeneity of PMDs, there is likely no unique cognitive profile. A study by Turconi and colleagues found that the non-verbal domain, notably the visuo-spatial ability, was more severely impaired. Working memory issues with verbal short-term memory were also noted [217]. Additionally, there are signs of autism spectrum disorders, and it has been suggested that mitochondrial malfunction may contribute to the illness process in autism spectrum disorders in general [218]. Generally speaking, psychiatric manifestations are common but often under-diagnosed in PMDs, including generalized anxiety, depression, and obsessive–compulsive spectrum disorder [219].

10. Discussion and Conclusions

The aim of this review was to describe the complexity of PMDs’ presentations and the existence of some peculiar features (“red flags”) that could help physicians on the diagnostic pathway; these are illustrated in Figure 1.
The peculiar symbiotic and semiautonomous nature of mitochondrial biology gives rise to a wide range of PMDs. For a genetic point of view, some typical phenotypes have been associated with specific genetic alterations. However, genotype–phenotype correlations, even for clinically defined syndromes, are complex and insufficient because of the genetic pleiotropy and the vast number of genes, either mitochondrial or nuclear-encoded, that may cause similar phenotypes. Moreover, while many patients fit into well-defined classic phenotypes, significant numbers are oligosymptomatic or have overlapping phenotypes. Generally, PMDs should be considered in any complex multisystem disorder, especially when neurological, eyes/ears, or endocrine manifestations are predominant. It is traditionally suggested that PMDs should be suspected in patients with an apparently unrelated involvement of two or more tissues.
If a PMD is suspected, family history is the first point to address; it must be taken meticulously, with special attention to minimal and apparently unspecific (“soft”) signs in the family. After the clinical assessment, neuroimaging, nerve conduction studies, electromyography, cardiac and retinal evaluation, as well as routine laboratory investigation, are important to support the diagnosis process and direct targeted molecular testing. In addition to phenotypic “red flags”, laboratory biomarkers are helpful for clinicians who suspect a PMD (Table 2). In recent years, specific guidelines for the diagnosis, management, and treatment of PMDs in pediatric patients and adults have been proposed [48,220,221]. The price of this diagnostic challenge is repaid by the large number of preclinical and clinical trials that have been developed, thanks to the implementation of national and international clinical registries, natural history studies and preclinical models. To date, therapies have been largely limited to supportive and symptomatic drugs, including vitamins and cofactors. However, small molecules approaches are emerging. They are principally focused on normalizing bioenergetic defects through different targets: modulation of oxidative stress, augmentation of mitochondrial biogenesis, regulation of autophagy, nucleotides pools restoration, restoration of the cellular NAD+ to NADH ratio, and others [222]. After idebenone for acute visual loss in LHON, only omaveloxolone for Friedreich’s ataxia have provided sufficient evidence for approval by U.S. Food and Drug administration [223]. Other promising therapeutics are allogenic stem-cell transplants, orthotopic liver transplantation, and enzyme replacement therapy in MNGIE, or deoxynucleoside supplementation therapy for TK2-deficient myopathy [138,224]. Gene therapy is another innovative approach. Allotopic AAV2-ND4 gene therapy for LHON patients harboring the m.11778G>A mtDNA mutation is one example [225]. In experimental models, significant progress has been achieved in the development of anti-sense peptide nucleic acids, endonucleases, transcription activator-like effector nuclease (TALENS), zinc finger nucleases, and CRISPR-Cas 9 that traverse the mitochondrial membrane and directly target mutant mtDNA for degradation [226].
In conclusion, to the non-specialist, PMDs are complicated and puzzling, difficult to recognize, and challenging to diagnose. However, important advances have been made in the field of PMDs diagnostics and phenotyping, as well as in the generation of pre-clinical models and therapeutics. Given the exciting era that mitochondrial medicine is experiencing, and also thanks to international community efforts, there is more need to rase awareness and stimulate interest towards these disorders.

Author Contributions

Conceptualization, P.L. and M.M.; methodology, P.L. and V.M. (Vincenzo Montano); validation, G.S., F.D. and M.M.; resources, G.S., F.D., C.B., V.M. (Vincenzo Micale), and M.M.; writing—original draft preparation, F.C., S.D.M. and P.L.; writing—review and editing, F.C., S.D.M. and P.L.; visualization, P.L.; supervision, P.L. and V.M. (Vincenzo Montano); project administration, M.M.; funding acquisition, M.M. All authors have read and agreed to the published version of the manuscript.

Funding

This work was partially supported by Telethon (grant GUP09004), Telethon-MITOCON (grant GSP16001), Ricerca finalizzata 2016 (02361495), European Joint Programme on Rare Diseases (EJPRD2019 project GENOMIT) and Italian Ministry of University and Research (2022B9WY4A).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We are grateful to the European Reference Networks NMD and RND (P.L., V.M. (Vincenzo Montano), G.S. and M.M.) as representatives for the Italian HCP partners.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ADOA: autosomal dominant optic atrophy; AHS: Alpers–Huttenlocher syndrome; ATP: adenosine triphosphate; BMI: body mass index; AUC: area under curve; CK: creatine kinase; CMT: Charcot–Marie–Tooth disease; CNS: central nervous system; COX: cytochrome c oxidase; cPEO: chronic progressive external ophthalmoplegia; CSF: cerebrospinal fluid; DNA: deoxyribonucleic acid; IOSCA: infantile-onset spinocerebellar ataxia; KSS: Kearns–Sayre syndrome; LHON: Leber’s hereditary optic neuropathy; LS: Leigh syndrome; MDSs: mitochondrial depletion syndromes; MELAS: mitochondrial encephalopathy with lactic acidosis and stroke-like episodes; MERRF: myoclonic epilepsy with ragged red fibers; MIDD: maternally inherited diabetes and deafness; MILS: maternally inherited Leigh syndrome; MIRAS: mitochondrial recessive ataxia syndrome; MNGIE: mitochondrial neurogastrointestinal encephalomyopathy; MRI: magnetic resonance imaging; MSL: multiple systemic lipomatosis; NARP: neuropathy, ataxia, and pigmentary retinopathy syndrome; OXPHOS: oxidative phosphorylation; PEO: progressive external ophthalmoplegia; PMDs: primary mitochondrial diseases; POLG: mitochondrial DNA polymerase gamma; RBFs: ragged blue fibers; RCGs: retinal ganglion cells; RNA: ribonucleic acid; RRFs: ragged red fibers; SANDO: sensory ataxia neuropathy dysarthria and ophthalmoplegia; SE: sensitivity; SLEs: stroke-like episodes; SP: specificity; tRNA: transfer ribonucleic acid.

References

  1. Gorman, G.S.; Schaefer, A.M.; Ng, Y.; Gomez, N.; Blakely, E.L.; Alston, C.L.; Feeney, C.; Horvath, R.; Yu-Wai-Man, P.; Chinnery, P.F.; et al. Prevalence of Nuclear and Mitochondrial DNA Mutations Related to Adult Mitochondrial Disease. Ann. Neurol. 2015, 77, 753–759. [Google Scholar] [CrossRef]
  2. Schlieben, L.D.; Prokisch, H. The Dimensions of Primary Mitochondrial Disorders. Front. Cell Dev. Biol. 2020, 8, 600079. [Google Scholar] [CrossRef]
  3. Ng, Y.S.; Bindoff, L.A.; Gorman, G.S.; Klopstock, T.; Kornblum, C.; Mancuso, M.; Mcfarland, R.; Sue, C.M.; Suomalainen, A.; Taylor, R.W.; et al. Mitochondrial Disease in Adults: Recent Advances and Future Promise. Lancet Neurol. 2021, 20, 573–584. [Google Scholar] [CrossRef] [PubMed]
  4. DiMauro, S.; Schon, E.A.; Carelli, V.; Hirano, M. The Clinical Maze of Mitochondrial Neurology. Nat. Rev. Neurol. 2013, 9, 429–444. [Google Scholar] [CrossRef] [PubMed]
  5. Payne, B.A.I.; Wilson, I.J.; Yu-Wai-Man, P.; Coxhead, J.; Deehan, D.; Horvath, R.; Taylor, R.W.; Samuels, D.C.; Santibanez-Koref, M.; Chinnery, P.F. Universal Heteroplasmy of Human Mitochondrial DNA. Hum. Mol. Genet. 2013, 22, 384–390. [Google Scholar] [CrossRef] [PubMed]
  6. McClelland, C.; Manousakis, G.; Lee, M.S. Progressive External Ophthalmoplegia. Curr. Neurol. Neurosci. Rep. 2016, 16, 53. [Google Scholar] [CrossRef] [PubMed]
  7. Smits, B.W.; Fermont, J.; Delnooz, C.C.S.; Kalkman, J.S.; Bleijenberg, G.; van Engelen, B.G.M. Disease Impact in Chronic Progressive External Ophthalmoplegia: More than Meets the Eye. Neuromuscul. Disord. 2011, 21, 272–278. [Google Scholar] [CrossRef]
  8. Smits, B.W.; Heijdra, Y.F.; Cuppen, F.W.A.; Van Engelen, B.G.M. Nature and Frequency of Respiratory Involvement in Chronic Progressive External Ophthalmoplegia. J. Neurol. 2011, 258, 2020–2025. [Google Scholar] [CrossRef]
  9. Orsucci, D.; Angelini, C.; Bertini, E.; Carelli, V.; Comi, G.P.; Federico, A.; Minetti, C.; Moggio, M.; Mongini, T.; Santorelli, F.M.; et al. Revisiting Mitochondrial Ocular Myopathies: A Study from the Italian Network. J. Neurol. 2017, 264, 1777–1784. [Google Scholar] [CrossRef]
  10. Orsucci, D.; Ienco, E.C.; Rossi, A.; Siciliano, G.; Mancuso, M. Mitochondrial Syndromes Revisited. J. Clin. Med. 2021, 10, 1249. [Google Scholar] [CrossRef]
  11. Soldath, P.; Madsen, K.L.; Buch, A.E.; Duno, M.; Wibrand, F.; Vissing, J. Pure Exercise Intolerance and Ophthalmoplegia Associated with the m.12,294G>A Mutation in the MT-TL2 Gene: A Case Report. BMC Musculoskelet. Disord. 2017, 18, 419. [Google Scholar] [CrossRef]
  12. Mancuso, M.; Angelini, C.; Bertini, E.; Carelli, V.; Comi, G.P.; Minetti, C.; Moggio, M.; Mongini, T.; Servidei, S.; Tonin, P.; et al. Fatigue and Exercise Intolerance in Mitochondrial Diseases. Literature Revision and Experience of the Italian Network of Mitochondrial Diseases. Neuromuscul. Disord. 2012, 22, S226–S229. [Google Scholar] [CrossRef]
  13. Distelmaier, F.; Klopstock, T. Neuroimaging in Mitochondrial Disease. In Handbook of Clinical Neurology; Elsevier: Amsterdam, The Netherlands, 2023; Volume 194, pp. 173–185. [Google Scholar] [CrossRef]
  14. Marques-Matos, C.; Reis, J.; Reis, C.; Castro, L.; Carvalho, M. Mitochondrial Encephalomyopathy With Lactic Acidosis and Strokelike Episodes Presenting Before 50 Years of Age. JAMA Neurol. 2016, 73, 604. [Google Scholar] [CrossRef] [PubMed]
  15. Goto, Y.; Horai, S.; Matsuoka, T.; Koga, Y.; Nihei, K.; Kobayashi, M.; Nonaka, I. Mitochondria1 Myopathy, Encephalopathy, Lactic Acidosis, and Stroke-like Episodes (MELAS): A Correlative Study of the Clinical Features and Mitochondrial DNA Mutation. Neurology 1992, 42, 545–550. [Google Scholar] [CrossRef] [PubMed]
  16. Ng, Y.S.; Bindoff, L.A.; Gorman, G.S.; Horvath, R.; Klopstock, T.; Mancuso, M.; Martikainen, M.H.; Mcfarland, R.; Nesbitt, V.; Pitceathly, R.D.S.; et al. Consensus-Based Statements for the Management of Mitochondrial Stroke-like Episodes. Wellcome Open Res. 2019, 4, 201. [Google Scholar] [CrossRef]
  17. Hikmat, O.; Naess, K.; Engvall, M.; Klingenberg, C.; Rasmussen, M.; Tallaksen, C.M.E.; Brodtkorb, E.; Ostergaard, E.; de Coo, I.F.M.; Pias-Peleteiro, L.; et al. Simplifying the Clinical Classification of Polymerase Gamma (POLG) Disease Based on Age of Onset; Studies Using a Cohort of 155 Cases. J. Inherit. Metab. Dis. 2020, 43, 726–736. [Google Scholar] [CrossRef] [PubMed]
  18. Hirano, M.; Ricci, E.; Richard Koenigsberger, M.; Defendini, R.; Pavlakis, S.G.; DeVivo, D.C.; DiMauro, S.; Rowland, L.P. MELAS: An Original Case and Clinical Criteria for Diagnosis. Neuromuscul. Disord. 1992, 2, 125–135. [Google Scholar] [CrossRef] [PubMed]
  19. Majamaa-Voltti, K.; Turkka, J.; Kortelainen, M.-L.; Huikuri, H.; Majamaa, K. Causes of Death in Pedigrees with the 3243A&gt;G Mutation in Mitochondrial DNA. J. Neurol. Neurosurg. Psychiatry 2008, 79, 209–211. [Google Scholar] [CrossRef] [PubMed]
  20. Ng, Y.S.; Lax, N.Z.; Blain, A.P.; Erskine, D.; Baker, M.R.; Polvikoski, T.; Thomas, R.H.; Morris, C.M.; Lai, M.; Whittaker, R.G.; et al. Forecasting Stroke-like Episodes and Outcomes in Mitochondrial Disease. Brain 2022, 145, 542–554. [Google Scholar] [CrossRef]
  21. Cheng, W.; Zhang, Y.; He, L. MRI Features of Stroke-Like Episodes in Mitochondrial Encephalomyopathy with Lactic Acidosis and Stroke-Like Episodes. Front. Neurol. 2022, 13, 843386. [Google Scholar] [CrossRef]
  22. Rahman, S. Leigh Syndrome. In Handbook of Clinical Neurology; Elsevier: Amsterdam, The Netherlands, 2023; Volume 194, pp. 43–63. [Google Scholar] [CrossRef]
  23. Green, D.R.; Kroemer, G. The Pathophysiology of Mitochondrial Cell Death. Science 2004, 305, 626–629. [Google Scholar] [CrossRef] [PubMed]
  24. Inak, G.; Rybak-Wolf, A.; Lisowski, P.; Pentimalli, T.M.; Jüttner, R.; Glažar, P.; Uppal, K.; Bottani, E.; Brunetti, D.; Secker, C.; et al. Defective Metabolic Programming Impairs Early Neuronal Morphogenesis in Neural Cultures and an Organoid Model of Leigh Syndrome. Nat. Commun. 2021, 12, 1929. [Google Scholar] [CrossRef]
  25. Sofou, K.; Steneryd, K.; Wiklund, L.M.; Tulinius, M.; Darin, N. MRI of the Brain in Childhood-Onset Mitochondrial Disorders with Central Nervous System Involvement. Mitochondrion 2013, 13, 364–371. [Google Scholar] [CrossRef] [PubMed]
  26. Hong, C.M.; Na, J.H.; Park, S.; Lee, Y.M. Clinical Characteristics of Early-Onset and Late-Onset Leigh Syndrome. Front. Neurol. 2020, 11, 267. [Google Scholar] [CrossRef] [PubMed]
  27. Martikainen, M.H.; Ng, Y.S.; Gorman, G.S.; Alston, C.L.; Blakely, E.L.; Schaefer, A.M.; Chinnery, P.F.; Burn, D.J.; Taylor, R.W.; McFarland, R.; et al. Clinical, Genetic, and Radiological Features of Extrapyramidal Movement Disorders in Mitochondrial Disease. JAMA Neurol. 2016, 73, 668–674. [Google Scholar] [CrossRef] [PubMed]
  28. Finsterer, J. Leigh and Leigh-Like Syndrome in Children and Adults. Pediatr. Neurol. 2008, 39, 223–235. [Google Scholar] [CrossRef] [PubMed]
  29. Bundschuh, F.A.; Hannappel, A.; Anderka, O.; Ludwig, B. Surf1, Associated with Leigh Syndrome in Humans, Is a Heme-Binding Protein in Bacterial Oxidase Biogenesis. J. Biol. Chem. 2009, 284, 25735–25741. [Google Scholar] [CrossRef]
  30. Ng, Y.S.; Martikainen, M.H.; Gorman, G.S.; Blain, A.; Bugiardini, E.; Bunting, A.; Schaefer, A.M.; Alston, C.L.; Blakely, E.L.; Sharma, S.; et al. Pathogenic Variants in MT-ATP6: A United Kingdom–Based Mitochondrial Disease Cohort Study. Ann. Neurol. 2019, 86, 310–315. [Google Scholar] [CrossRef]
  31. Ng, Y.S.; Lax, N.Z.; Maddison, P.; Alston, C.L.; Blakely, E.L.; Hepplewhite, P.D.; Riordan, G.; Meldau, S.; Chinnery, P.F.; Pierre, G.; et al. MT-ND5 Mutation Exhibits Highly Variable Neurological Manifestations at Low Mutant Load. EBioMedicine 2018, 30, 86–93. [Google Scholar] [CrossRef]
  32. La Morgia, C.; Caporali, L.; Gandini, F.; Olivieri, A.; Toni, F.; Nassetti, S.; Brunetto, D.; Stipa, C.; Scaduto, C.; Parmeggiani, A.; et al. Association of the MtDNA m.4171C&gt;A/MT-ND1 Mutation with Both Optic Neuropathy and Bilateral Brainstem Lesions. BMC Neurol. 2014, 14, 116. [Google Scholar] [CrossRef]
  33. Nakagawa, E.; Hirano, S.; Yamanouchi, H.; Goto, Y.; Nonaka, I.; Takashima, S. Progressive Brainstem and White Matter Lesions in Kearns—Sayre Syndrome: A Case Report. Brain Dev. 1994, 16, 416–418. [Google Scholar] [CrossRef] [PubMed]
  34. Chu, B.C.; Terae, S.; Takahashi, C.; Kikuchi, Y.; Miyasaka, K.; Abe, S.; Minowa, K.; Sawamura, T. MRI of the Brain in the Kearns-Sayre Syndrome: Report of Four Cases and a Review. Neuroradiology 1999, 41, 759–764. [Google Scholar] [CrossRef]
  35. Hai, N.T.; Ngan, V.K.; Giang, N.Q.; Hoang, V.H.; Viet, L.A.; Duc, N.T.; Hien, T.T.T.; He, D.V.; Duc, N.M. The Magnetic Resonance Imaging of Leigh Syndrome in a Child. Clin. Ter. 2021, 172, 500–503. [Google Scholar] [CrossRef]
  36. Wesół-Kucharska, D.; Rokicki, D.; Jezela-Stanek, A. Epilepsy in Mitochondrial Diseases—Current State of Knowledge on Aetiology and Treatment. Children 2021, 8, 532. [Google Scholar] [CrossRef]
  37. Patel, K.P.; O’Brien, T.W.; Subramony, S.H.; Shuster, J.; Stacpoole, P.W. The Spectrum of Pyruvate Dehydrogenase Complex Deficiency: Clinical, Biochemical and Genetic Features in 371 Patients. Mol. Genet. Metab. 2012, 106, 385–394. [Google Scholar] [CrossRef]
  38. Chang, X.; Wu, Y.; Zhou, J.; Meng, H.; Zhang, W.; Guo, J. A Meta-Analysis and Systematic Review of Leigh Syndrome: Clinical Manifestations, Respiratory Chain Enzyme Complex Deficiency, and Gene Mutations. Medicine 2020, 99, E18634. [Google Scholar] [CrossRef] [PubMed]
  39. Finsterer, J.; Zarrouk-Mahjoub, S. Management of Epilepsy in MERRF Syndrome. Seizure 2017, 50, 166–170. [Google Scholar] [CrossRef] [PubMed]
  40. El-Hattab, A.W.; Adesina, A.M.; Jones, J.; Scaglia, F. MELAS Syndrome: Clinical Manifestations, Pathogenesis, and Treatment Options. Mol. Genet. Metab. 2015, 116, 4–12. [Google Scholar] [CrossRef] [PubMed]
  41. Lim, A.; Thomas, R.H. The Mitochondrial Epilepsies. Eur. J. Paediatr. Neurol. 2020, 24, 47–52. [Google Scholar] [CrossRef] [PubMed]
  42. Surana, S.; Rossor, T.; Hassell, J.; Boyd, S.; D’Arco, F.; Aylett, S.; Bhate, S.; Carr, L.; Das, K.; DeVile, C.; et al. Diagnostic Algorithm for Children Presenting with Epilepsia Partialis Continua. Epilepsia 2020, 61, 2224–2233. [Google Scholar] [CrossRef]
  43. Lopriore, P.; Gomes, F.; Montano, V.; Siciliano, G.; Mancuso, M. Mitochondrial Epilepsy, a Challenge for Neurologists. Int. J. Mol. Sci. 2022, 23, 13216. [Google Scholar] [CrossRef] [PubMed]
  44. Anagnostou, M.E.; Ng, Y.S.; Taylor, R.W.; McFarland, R. Epilepsy Due to Mutations in the Mitochondrial Polymerase Gamma (POLG) Gene: A Clinical and Molecular Genetic Review. Epilepsia 2016, 57, 1531–1545. [Google Scholar] [CrossRef] [PubMed]
  45. Hikmat, O.; Eichele, T.; Tzoulis, C.; Bindoff, L. Understanding the Epilepsy in POLG Related Disease. Int. J. Mol. Sci. 2017, 18, 1845. [Google Scholar] [CrossRef] [PubMed]
  46. Rahman, S. Mitochondrial Diseases and Status Epilepticus. Epilepsia 2018, 59, 70–77. [Google Scholar] [CrossRef] [PubMed]
  47. Shorvon, S.; Ferlisi, M. The Treatment of Super-Refractory Status Epilepticus: A Critical Review of Available Therapies and a Clinical Treatment Protocol. Brain 2011, 134, 2802–2818. [Google Scholar] [CrossRef] [PubMed]
  48. Parikh, S.; Goldstein, A.; Karaa, A.; Koenig, M.K.; Anselm, I.; Brunel-Guitton, C.; Christodoulou, J.; Cohen, B.H.; Dimmock, D.; Enns, G.M.; et al. Patient Care Standards for Primary Mitochondrial Disease: A Consensus Statement from the Mitochondrial Medicine Society. Genet. Med. 2017, 19, 1380–1397. [Google Scholar] [CrossRef]
  49. Rahman, S. Advances in the Treatment of Mitochondrial Epilepsies. Epilepsy Behav. 2019, 101, 106546. [Google Scholar] [CrossRef] [PubMed]
  50. Ticci, C.; Sicca, F.; Ardissone, A.; Bertini, E.; Carelli, V.; Diodato, D.; Di Vito, L.; Filosto, M.; La Morgia, C.; Lamperti, C.; et al. Mitochondrial Epilepsy: A Cross-Sectional Nationwide Italian Survey. Neurogenetics 2020, 21, 87–96. [Google Scholar] [CrossRef]
  51. Lee, S.; Na, J.H.; Lee, Y.M. Epilepsy in Leigh Syndrome with Mitochondrial DNA Mutations. Front. Neurol. 2019, 10, 496. [Google Scholar] [CrossRef]
  52. Lopriore, P.; Ricciarini, V.; Siciliano, G.; Mancuso, M.; Montano, V. Mitochondrial Ataxias: Molecular Classification and Clinical Heterogeneity. Neurol. Int. 2022, 14, 337–356. [Google Scholar] [CrossRef]
  53. Vergara, R.C.; Jaramillo-Riveri, S.; Luarte, A.; Moënne-Loccoz, C.; Fuentes, R.; Couve, A.; Maldonado, P.E. The Energy Homeostasis Principle: Neuronal Energy Regulation Drives Local Network Dynamics Generating Behavior. Front. Comput. Neurosci. 2019, 13, 49. [Google Scholar] [CrossRef]
  54. Tranchant, C.; Anheim, M. Movement Disorders in Mitochondrial Diseases. Rev. Neurol. 2016, 172, 524–529. [Google Scholar] [CrossRef]
  55. Filosto, M.; Mancuso, M. Mitochondrial Diseases: A Nosological Update. Acta Neurol. Scand. 2007, 115, 211–221. [Google Scholar] [CrossRef]
  56. Hakonen, A.H.; Heiskanen, S.; Juvonen, V.; Lappalainen, I.; Luoma, P.T.; Rantamäki, M.; Van Goethem, G.; Löfgren, A.; Hackman, P.; Paetau, A.; et al. Mitochondrial DNA Polymerase W748S Mutation: A Common Cause of Autosomal Recessive Ataxia with Ancient European Origin. Am. J. Hum. Genet. 2005, 77, 430–441. [Google Scholar] [CrossRef]
  57. Schicks, J.; Synofzik, M.; Schulte, C.; Schöls, L. POLG, but Not PEO1, Is a Frequent Cause of Cerebellar Ataxia in Central Europe. Mov. Disord. 2010, 25, 2678–2682. [Google Scholar] [CrossRef] [PubMed]
  58. Goffart, S.; Cooper, H.M.; Tyynismaa, H.; Wanrooij, S.; Suomalainen, A.; Spelbrink, J.N. Twinkle Mutations Associated with Autosomal Dominant Progressive External Ophthalmoplegia Lead to Impaired Helicase Function and in Vivo MtDNA Replication Stalling. Hum. Mol. Genet. 2009, 18, 328–340. [Google Scholar] [CrossRef] [PubMed]
  59. Fadic, R.; Russell, J.A.; Vedanarayanan, V.V.; Lehar, M.; Kuncl, R.W.; Johns, D.R. Sensory Ataxic Neuropathy as the Presenting Feature of a Novel Mitochondrial Disease. Neurology 1997, 49, 239–245. [Google Scholar] [CrossRef] [PubMed]
  60. Brunetti, D.; Torsvik, J.; Dallabona, C.; Teixeira, P.; Sztromwasser, P.; Fernandez-Vizarra, E.; Cerutti, R.; Reyes, A.; Preziuso, C.; D’Amati, G.; et al. Defective PITRM1 Mitochondrial Peptidase Is Associated with Aβ Amyloidotic Neurodegeneration. EMBO Mol. Med. 2016, 8, 176–190. [Google Scholar] [CrossRef]
  61. Kiferle, L.; Orsucci, D.; Mancuso, M.; Lo Gerfo, A.; Petrozzi, L.; Siciliano, G.; Ceravolo, R.; Bonuccelli, U. Twinkle Mutation in an Italian Family with External Progressive Ophthalmoplegia and Parkinsonism: A Case Report and an Update on the State of Art. Neurosci. Lett. 2013, 556, 1–4. [Google Scholar] [CrossRef]
  62. Horvath, R.; Kley, R.A.; Vorgerd, M. Parkinson Syndrome, Neuropathy, and Myopathy Caused by the Mutation A8344G (MERRF) in TRNA Lys. Neurology 2007, 68, 56–58. [Google Scholar] [CrossRef]
  63. Dermaut, B.; Seneca, S.; Dom, L.; Smets, K.; Ceulemans, L.; Smet, J.; De Paepe, B.; Tousseyn, S.; Weckhuysen, S.; Gewillig, M.; et al. Progressive Myoclonic Epilepsy as an Adult-Onset Manifestation of Leigh Syndrome Due to m.14487T>C. J. Neurol. Neurosurg. Psychiatry 2010, 81, 90–93. [Google Scholar] [CrossRef] [PubMed]
  64. Thobois, S.; Vighetto, A.; Grochowicki, M.; Godinot, C.; Broussolle, E.; Aimard, G. [Leber “plus” Disease: Optic Neuropathy, Parkinsonian Syndrome and Supranuclear Ophthalmoplegia]. Rev. Neurol. 1997, 153, 595–598. [Google Scholar] [PubMed]
  65. De Coo, I.F.M.; Renier, W.O.; Ruitenbeek, W.; Ter Laak, H.J.; Bakker, M.; Schägger, H.; Van Oost, B.A.; Smeets, H.J.M. A 4–Base Pair Deletion in the Mitochondrial Cytochrome b Gene Associated with Parkinsonism/MELAS Overlap Syndrome. Ann. Neurol. 1999, 45, 130–133. [Google Scholar] [CrossRef] [PubMed]
  66. Montano, V.; Orsucci, D.; Carelli, V.; La Morgia, C.; Valentino, M.L.; Lamperti, C.; Marchet, S.; Musumeci, O.; Toscano, A.; Primiano, G.; et al. Adult-Onset Mitochondrial Movement Disorders: A National Picture from the Italian Network. J. Neurol. 2022, 269, 1413–1421. [Google Scholar] [CrossRef]
  67. Yorns, W.R.; Hardison, H.H. Mitochondrial Dysfunction in Migraine. Semin. Pediatr. Neurol. 2013, 20, 188–193. [Google Scholar] [CrossRef]
  68. Vollono, C.; Primiano, G.; Della Marca, G.; Losurdo, A.; Servidei, S. Migraine in Mitochondrial Disorders: Prevalence and Characteristics. Cephalalgia 2018, 38, 1093–1106. [Google Scholar] [CrossRef]
  69. Nesbitt, V.; Pitceathly, R.D.S.; Turnbull, D.M.; Taylor, R.W.; Sweeney, M.G.; Mudanohwo, E.E.; Rahman, S.; Hanna, M.G.; McFarland, R. The UK MRC Mitochondrial Disease Patient Cohort Study: Clinical Phenotypes Associated with the m.3243A>G Mutation—Implications for Diagnosis and Management. J. Neurol. Neurosurg. Psychiatry 2013, 84, 936–938. [Google Scholar] [CrossRef]
  70. Mancuso, M.; Orsucci, D.; Angelini, C.; Bertini, E.; Carelli, V.; Comi, G.P.; Donati, A.; Minetti, C.; Moggio, M.; Mongini, T.; et al. The m.3243A>G Mitochondrial DNA Mutation and Related Phenotypes. A Matter of Gender? J. Neurol. 2014, 261, 504–510. [Google Scholar] [CrossRef]
  71. Primiano, G.; Rollo, E.; Romozzi, M.; Calabresi, P.; Servidei, S.; Vollono, C. Preventive Migraine Treatment in Mitochondrial Diseases: A Case Report of Erenumab Efficacy and Literature Review. Neurol. Sci. 2022, 43, 6955–6959. [Google Scholar] [CrossRef]
  72. Pareyson, D.; Piscosquito, G.; Moroni, I.; Salsano, E.; Zeviani, M. Peripheral Neuropathy in Mitochondrial Disorders. Lancet Neurol. 2013, 12, 1011–1024. [Google Scholar] [CrossRef]
  73. Finsterer, J. Mitochondrial Neuropathy. Clin. Neurol. Neurosurg. 2005, 107, 181–186. [Google Scholar] [CrossRef]
  74. Garone, C.; Tadesse, S.; Hirano, M. Clinical and Genetic Spectrum of Mitochondrial Neurogastrointestinal Encephalomyopathy. Brain 2011, 134, 3326–3332. [Google Scholar] [CrossRef]
  75. Gilhuis, H.J.; Schelhaas, H.J.; Cruysberg, J.R.M.; Zwarts, M.J. Demyelinating Polyneuropathy in Leber Hereditary Optic Neuropathy. Neuromuscul. Disord. 2006, 16, 394–395. [Google Scholar] [CrossRef]
  76. Santoro, L.; Carrozzo, R.; Malandrini, A.; Piemonte, F.; Patrono, C.; Villanova, M.; Tessa, A.; Palmeri, S.; Bertini, E.; Santorelli, F.M. A Novel SURF1 Mutation Results in Leigh Syndrome with Peripheral Neuropathy Caused by Cytochrome c Oxidase Deficiency. Neuromuscul. Disord. 2000, 10, 450–453. [Google Scholar] [CrossRef] [PubMed]
  77. Bouillot, S.; Martin-Negrier, M.L.; Vital, A.; Ferrer, X.; Lagueny, A.; Vincent, D.; Coquet, M.; Orgogozo, J.M.; Bloch, B.; Vital, C. Peripheral Neuropathy Associated with Mitochondrial Disorders: 8 Cases and Review of the Literature. J. Peripher. Nerv. Syst. 2002, 7, 213–220. [Google Scholar] [CrossRef] [PubMed]
  78. Rusanen, H.; Majamaa, K.; Tolonen, U.; Remes, A.M.; Myllyla, R.; Hassinen, I.E. Demyelinating Polyneuropathy in a Patient with the TRNALeu(Uur) Mutation at Base Pair 3243 of the Mitochondrial DNA. Neurology 1995, 45, 1188–1192. [Google Scholar] [CrossRef] [PubMed]
  79. Rahman, S.; Hanna, M.G. Diagnosis and Therapy in Neuromuscular Disorders: Diagnosis and New Treatments in Mitochondrial Diseases. J. Neurol. Neurosurg. Psychiatry 2009, 80, 943–953. [Google Scholar] [CrossRef] [PubMed]
  80. Verhoeven, K. MFN2 Mutation Distribution and Genotype/Phenotype Correlation in Charcot-Marie-Tooth Type 2. Brain 2006, 129, 2093–2102. [Google Scholar] [CrossRef]
  81. Nelis, E.; Van Broeckhoven, C. Estimation of the Mutation Frequencies in Charcot-Marie-Tooth Disease Type 1 and Hereditary Neuropathy with Liability to Pressure Palsies: A European Collaborative Study. Eur. J. Hum. Genet. 1996, 4, 25–33. [Google Scholar] [CrossRef]
  82. Horvath, R.; Medina, J.; Reilly, M.M.; Shy, M.E.; Zuchner, S. Peripheral Neuropathy in Mitochondrial Disease. In Handbook of Clinical Neurology; Elsevier: Amsterdam, The Netherlands, 2023; pp. 99–116. [Google Scholar] [CrossRef]
  83. Saporta, A.S.D.; Sottile, S.L.; Miller, L.J.; Feely, S.M.E.; Siskind, C.E.; Shy, M.E. Charcot-Marie-Tooth Disease Subtypes and Genetic Testing Strategies. Ann. Neurol. 2011, 69, 22–33. [Google Scholar] [CrossRef]
  84. Murphy, S.M.; Laura, M.; Fawcett, K.; Pandraud, A.; Liu, Y.T.; Davidson, G.L.; Rossor, A.M.; Polke, J.M.; Castleman, V.; Manji, H.; et al. Charcot-Marie-Tooth Disease: Frequency of Genetic Subtypes and Guidelines for Genetic Testing. J. Neurol. Neurosurg. Psychiatry 2012, 83, 706–710. [Google Scholar] [CrossRef]
  85. Saporta, M.A.; Dang, V.; Volfson, D.; Zou, B.; Xie, X.S.; Adebola, A.; Liem, R.K.; Shy, M.; Dimos, J.T. Axonal Charcot-Marie-Tooth Disease Patient-Derived Motor Neurons Demonstrate Disease-Specific Phenotypes Including Abnormal Electrophysiological Properties. Exp. Neurol. 2015, 263, 190–199. [Google Scholar] [CrossRef] [PubMed]
  86. Sharma, C.; Kim, S.; Nam, Y.; Jung, U.J.; Kim, S.R. Mitochondrial Dysfunction as a Driver of Cognitive Impairment in Alzheimer’s Disease. Int. J. Mol. Sci. 2021, 22, 4850. [Google Scholar] [CrossRef]
  87. Moore, H.L.; Kelly, T.; Bright, A.; Field, R.H.; Schaefer, A.M.; Blain, A.P.; Taylor, R.W.; McFarland, R.; Turnbull, D.M.; Gorman, G.S. Cognitive Deficits in Adult m.3243A>G- and m.8344A>G-Related Mitochondrial Disease: Importance of Correcting for Baseline Intellectual Ability. Ann. Clin. Transl. Neurol. 2019, 6, 826–836. [Google Scholar] [CrossRef]
  88. Moore, H.L.; Blain, A.P.; Turnbull, D.M.; Gorman, G.S. Systematic Review of Cognitive Deficits in Adult Mitochondrial Disease. Eur. J. Neurol. 2020, 27, 3–17. [Google Scholar] [CrossRef] [PubMed]
  89. Lang, C.J.G.; Brenner, P.; HeuB, D.; Engelhardt, A.; Reichmann, H.; Seibel, P.; Neundörfer, B. Neuropsychological Status of Mitochondrial Encephalomyopathies. Eur. J. Neurol. 1995, 2, 171–176. [Google Scholar] [CrossRef] [PubMed]
  90. Kaufmann, P.; Engelstad, K.; Wei, B.Y.; Kulikova, R.; Oskoui, M.; Sproule, D.; Battista, V.; Koenigsberger, C.D.; Pascual, P.J.; Shanske, S.; et al. Natural History of MELAS Associated with Mitochondrial DNA m.3243AG Genotype. Neurology 2011, 77, 1965–1971. [Google Scholar] [CrossRef]
  91. Nomura, T.; Ota, M.; Kotake, N.; Tanaka, K. [Two Cases of MERRF (Myoclonus Epilepsy Associated with Ragged Red Fibers) Showing Different Clinical Features in the Same Family]. Rinsho Shinkeigaku 1993, 33, 1198–1200. [Google Scholar]
  92. Kalimo, H.; Lundberg, P.O.; Olsson, Y. Familial Subacute Necrotizing Encephalomyelopathy of the Adult Form (Adult Leigh Syndrome). Ann. Neurol. 1979, 6, 200–206. [Google Scholar] [CrossRef]
  93. Tsao, C.-Y.; Mendell, J.R.; Bartholomew, D. High Mitochondrial DNA T8993G Mutation (>90%) Without Typical Features of Leigh’s and NARP Syndromes. J. Child. Neurol. 2001, 16, 533–535. [Google Scholar] [CrossRef]
  94. McFarland, R.; Taylor, R.W.; Turnbull, D.M. The Neurology of Mitochondrial DNA Disease. Lancet Neurol. 2002, 1, 343–351. [Google Scholar] [CrossRef] [PubMed]
  95. Lascaratos, G.; Garway-Heath, D.F.; Willoughby, C.E.; Chau, K.Y.; Schapira, A.H.V. Mitochondrial Dysfunction in Glaucoma: Understanding Genetic Influences. Mitochondrion 2012, 12, 202–212. [Google Scholar] [CrossRef] [PubMed]
  96. Majander, A.; Robson, A.G.; João, C.; Holder, G.E.; Chinnery, P.F.; Moore, A.T.; Votruba, M.; Stockman, A.; Yu-Wai-Man, P. The Pattern of Retinal Ganglion Cell Dysfunction in Leber Hereditary Optic Neuropathy. Mitochondrion 2017, 36, 138–149. [Google Scholar] [CrossRef]
  97. Marotta, R.; Chin, J.; Chiotis, M.; Shuey, N.; Collins, S.J. Long-Term Screening for Primary Mitochondrial DNA Variants Associated with Leber Hereditary Optic Neuropathy: Incidence, Penetrance and Clinical Features. Mitochondrion 2020, 54, 128–132. [Google Scholar] [CrossRef]
  98. Boggan, R.M.; Lim, A.; Taylor, R.W.; McFarland, R.; Pickett, S.J. Resolving Complexity in Mitochondrial Disease: Towards Precision Medicine. Mol. Genet. Metab. 2019, 128, 19–29. [Google Scholar] [CrossRef]
  99. Kirkman, M.A.; Yu-Wai-Man, P.; Korsten, A.; Leonhardt, M.; Dimitriadis, K.; De Coo, I.F.; Klopstock, T.; Chinnery, P.F. Geneenvironment Interactions in Leber Hereditary Optic Neuropathy. Brain 2009, 132, 2317–2326. [Google Scholar] [CrossRef]
  100. Yu-Wai-Man, P.; Chinnery, P.F. Dominant Optic Atrophy: Novel OPA1 Mutations and Revised Prevalence Estimates. Ophthalmology 2013, 120, 1712–1712.e1. [Google Scholar] [CrossRef]
  101. Borrelli, E.; Triolo, G.; Cascavilla, M.L.; La Morgia, C.; Rizzo, G.; Savini, G.; Balducci, N.; Nucci, P.; Giglio, R.; Darvizeh, F.; et al. Changes in Choroidal Thickness Follow the RNFL Changes in Leber’s Hereditary Optic Neuropathy. Sci. Rep. 2016, 6, 37332. [Google Scholar] [CrossRef] [PubMed]
  102. Newman, N.J.; Carelli, V.; Taiel, M.; Yu-Wai-Man, P. Visual Outcomes in Leber Hereditary Optic Neuropathy Patients with the m.11778G>A (MTND4) Mitochondrial DNA Mutation. J. Neuro-Ophthalmol. 2020, 40, 547–557. [Google Scholar] [CrossRef] [PubMed]
  103. Zhang, J.; Ji, Y.; Chen, J.; Xu, M.; Wang, G.; Ci, X.; Lin, B.; Mo, J.Q.; Zhou, X.; Guan, M.X. Assocation between Leber’s Hereditary Optic Neuropathy and MT-ND1 3460G>a Mutation-Induced Alterations in Mitochondrial Function, Apoptosis, and Mitophagy. Investig. Ophthalmol. Vis. Sci. 2021, 62, 38. [Google Scholar] [CrossRef]
  104. Zhang, J.; Zhao, F.; Fu, Q.; Liang, M.; Tong, Y.; Liu, X.; Lin, B.; Mi, H.; Zhang, M.; Wei, Q.P.; et al. Mitochondrial Haplotypes May Modulate the Phenotypic Manifestation of the LHON-Associated m.14484T>C (MT-ND6) Mutation in Chinese Families. Mitochondrion 2013, 13, 772–781. [Google Scholar] [CrossRef]
  105. d’Almeida, O.C.; Violante, I.R.; Quendera, B.; Castelo-Branco, M. Mitochondrial Pathophysiology beyond the Retinal Ganglion Cell: Occipital GABA Is Decreased in Autosomal Dominant Optic Neuropathy. Graefe’s Arch. Clin. Exp. Ophthalmol. 2018, 256, 2341–2348. [Google Scholar] [CrossRef]
  106. Yu-Wai-Man, P.; Griffiths, P.G.; Gorman, G.S.; Lourenco, C.M.; Wright, A.F.; Auer-Grumbach, M.; Toscano, A.; Musumeci, O.; Valentino, M.L.; Caporali, L.; et al. Multi-System Neurological Disease Is Common in Patients with OPA1 Mutations. Brain 2010, 133, 771–786. [Google Scholar] [CrossRef]
  107. Isashiki, Y.; Nakagawa, M.; Ohba, N.; Kamimura, K.; Sakoda, Y.; Higuchi, I.; Izumo, S.; Osame, M. Retinal Manifestations in Mitochondrial Diseases Associated with Mitochondrial DNA Mutation. Acta Ophthalmol. Scand. 1998, 76, 6–13. [Google Scholar] [CrossRef] [PubMed]
  108. Stamate, A.C.; Burcea, M.; Zemba, M. Unilateral pigmentary retinopathy—A review of literature and case presentation. Rom. J. Ophthalmol. 2016, 60, 47–52. [Google Scholar] [PubMed]
  109. Shemesh, A.; Margolin, E. Kearns-Sayre Syndrome. In StatPearls [Internet]; StatPearls Publishing: Treasure Island, FL, USA, 2023. [Google Scholar]
  110. Latvala, T.; Mustonen, E.; Uusitalo, R.; Majama, K. Pigmentary Retinopathy in Patients with the Melas Mutation 3243A→G in Mitochondrial DNA. Graefe’s Arch. Clin. Exp. Ophthalmol. 2002, 240, 795–801. [Google Scholar] [CrossRef]
  111. Han, J.; Lee, Y.M.; Kim, S.M.; Han, S.Y.; Lee, J.B.; Han, S.H. Ophthalmological Manifestations in Patients with Leigh Syndrome. Br. J. Ophthalmol. 2015, 99, 528–535. [Google Scholar] [CrossRef] [PubMed]
  112. Smith, P.R.; Bain, S.C.; Good, P.A.; Hattersley, A.T.; Barnett, A.H.; Gibson, J.M.; Dodson, P.M. Pigmentary Retinal Dystrophy and the Syndrome of Maternally Inherited Diabetes and Deafness Caused by the Mitochondrial DNA 3243 TRNA Leu A to G Mutation. Ophthalmology 1999, 106, 1101–1108. [Google Scholar] [CrossRef] [PubMed]
  113. Al-Enezi, M.; Al-Saleh, H.; Nasser, M. Mitochondrial Disorders with Significant Ophthalmic Manifestations. Middle East. Afr. J. Ophthalmol. 2008, 15, 81. [Google Scholar] [CrossRef]
  114. Kullar, P.J.; Quail, J.; Lindsey, P.; Wilson, J.A.; Horvath, R.; Yu-Wai-Man, P.; Gorman, G.S.; Taylor, R.W.; Ng, Y.; McFarland, R.; et al. Both Mitochondrial DNA and Mitonuclear Gene Mutations Cause Hearing Loss through Cochlear Dysfunction. Brain 2016, 139, e33. [Google Scholar] [CrossRef]
  115. Santarelli, R.; Rossi, R.; Scimemi, P.; Cama, E.; Valentino, M.L.; La Morgia, C.; Caporali, L.; Liguori, R.; Magnavita, V.; Monteleone, A.; et al. OPA1-Related Auditory Neuropathy: Site of Lesion and Outcome of Cochlear Implantation. Brain 2015, 138, 563–576. [Google Scholar] [CrossRef] [PubMed]
  116. Liu, K.; Sun, W.; Zhou, X.; Bao, S.; Gong, S.; He, D.Z. Editorial: Hearing Loss and Cognitive Disorders. Front. Neurosci. 2022, 16, 902405. [Google Scholar] [CrossRef]
  117. Zou, T.; Ye, B.; Chen, K.; Zhang, A.; Guo, D.; Pan, Y.; Ding, R.; Hu, H.; Sun, X.; Xiang, M. Impacts of Impaired Mitochondrial Dynamics in Hearing Loss: Potential Therapeutic Targets. Front. Neurosci. 2022, 16, 998507. [Google Scholar] [CrossRef] [PubMed]
  118. Sharma, N.; Kumari, D.; Panigrahi, I.; Khetarpal, P. A Systematic Review of the Monogenic Causes of Non-Syndromic Hearing Loss (NSHL) and Discussion of Current Diagnosis and Treatment Options. Clin. Genet. 2023, 103, 16–34. [Google Scholar] [CrossRef] [PubMed]
  119. Orsucci, D.; Caldarazzo Ienco, E.; Siciliano, G.; Mancuso, M. Mitochondrial Disorders and Drugs: What Every Physician Should Know. Drugs Context. 2019, 8, 212588. [Google Scholar] [CrossRef] [PubMed]
  120. Ayers, M.; Horslen, S.P.; Gómez, A.M.; Squires, J.E. Mitochondrial Hepatopathy. Clin. Liver. Dis. 2022, 26, 421–438. [Google Scholar] [CrossRef]
  121. Grattagliano, I.; Russmann, S.; Diogo, C.; Bonfrate, L.; Oliveira, P.J.; Wang, D.Q.-H.; Portincasa, P. Mitochondria in Chronic Liver Disease. Curr. Drug Targets 2011, 12, 879–893. [Google Scholar] [CrossRef]
  122. Schara, U.; Von Kleist-Retzow, J.C.; Lainka, E.; Gerner, P.; Pyle, A.; Smith, P.M.; Lochmüller, H.; Czermin, B.; Abicht, A.; Holinski-Feder, E.; et al. Acute Liver Failure with Subsequent Cirrhosis as the Primary Manifestation of TRMU Mutations. J. Inherit. Metab. Dis. 2011, 34, 197–201. [Google Scholar] [CrossRef]
  123. Horvath, R.; Hudson, G.; Ferrari, G.; Fütterer, N.; Ahola, S.; Lamantea, E.; Prokisch, H.; Lochmüller, H.; McFarland, R.; Ramesh, V.; et al. Phenotypic Spectrum Associated with Mutations of the Mitochondrial Polymerase γ Gene. Brain 2006, 129, 1674–1684. [Google Scholar] [CrossRef]
  124. Saneto, R.P.; Cohen, B.H.; Copeland, W.C.; Naviaux, R.K. Alpers-Huttenlocher Syndrome. Pediatr. Neurol. 2013, 48, 167–178. [Google Scholar] [CrossRef]
  125. De Vries, M.C.; Brown, D.A.; Allen, M.E.; Bindoff, L.; Gorman, G.S.; Karaa, A.; Keshavan, N.; Lamperti, C.; McFarland, R.; Ng, Y.S.; et al. Safety of Drug Use in Patients with a Primary Mitochondrial Disease: An International Delphi-Based Consensus. J. Inherit. Metab. Dis. 2020, 43, 800–818. [Google Scholar] [CrossRef]
  126. Hakonen, A.H.; Isohanni, P.; Paetau, A.; Herva, R.; Suomalainen, A.; Lönnqvist, T. Recessive Twinkle Mutations in Early Onset Encephalopathy with MtDNA Depletion. Brain 2007, 130, 3032–3040. [Google Scholar] [CrossRef]
  127. Spinazzola, A.; Invernizzi, F.; Carrara, F.; Lamantea, E.; Donati, A.; Dirocco, M.; Giordano, I.; Meznaric-Petrusa, M.; Baruffini, E.; Ferrero, I.; et al. Clinical and Molecular Features of Mitochondrial DNA Depletion Syndromes. J. Inherit. Metab. Dis. 2009, 32, 143–158. [Google Scholar] [CrossRef] [PubMed]
  128. Dalla Rosa, I.; Cámara, Y.; Durigon, R.; Moss, C.F.; Vidoni, S.; Akman, G.; Hunt, L.; Johnson, M.A.; Grocott, S.; Wang, L.; et al. MPV17 Loss Causes Deoxynucleotide Insufficiency and Slow DNA Replication in Mitochondria. PLoS Genet. 2016, 12, e1005779. [Google Scholar] [CrossRef] [PubMed]
  129. Tadiboyina, V.T.; Rupar, A.; Atkison, P.; Feigenbaum, A.; Kronick, J.; Wang, J.; Hegele, R.A. Novel Mutation in DGUOK in Hepatocerebral Mitochondrial DNA Depletion Syndrome Associated with Cystathioninuria. Am. J. Med. Genet. A 2005, 135, 289–291. [Google Scholar] [CrossRef] [PubMed]
  130. Dimmock, D.P.; Zhang, Q.; Dionisi-Vici, C.; Carrozzo, R.; Shieh, J.; Tang, L.-Y.; Truong, C.; Schmitt, E.; Sifry-Platt, M.; Lucioli, S.; et al. Clinical and Molecular Features of Mitochondrial DNA Depletion Due to Mutations in Deoxyguanosine Kinase. Hum. Mutat. 2008, 29, 330–331. [Google Scholar] [CrossRef] [PubMed]
  131. Uusimaa, J.; Evans, J.; Smith, C.; Butterworth, A.; Craig, K.; Ashley, N.; Liao, C.; Carver, J.; Diot, A.; Macleod, L.; et al. Clinical, Biochemical, Cellular and Molecular Characterization of Mitochondrial DNA Depletion Syndrome Due to Novel Mutations in the MPV17 Gene. Eur. J. Hum. Genet. 2014, 22, 184–191. [Google Scholar] [CrossRef] [PubMed]
  132. Tiranti, V.; Viscomi, C.; Hildebrandt, T.; Di Meo, I.; Mineri, R.; Tiveron, C.; Levitt, M.D.; Prelle, A.; Fagiolari, G.; Rimoldi, M.; et al. Loss of ETHE1, a Mitochondrial Dioxygenase, Causes Fatal Sulfide Toxicity in Ethylmalonic Encephalopathy. Nat. Med. 2009, 15, 200–205. [Google Scholar] [CrossRef]
  133. Chen, X.; Han, L.; Yao, H. Novel Compound Heterozygous Variants of ETHE1 Causing Ethylmalonic Encephalopathy in a Chinese Patient: A Case Report. Front. Genet. 2020, 11, 341. [Google Scholar] [CrossRef]
  134. Hassan, S.; Mahmoud, A.; Mohammed, T.O.; Mohammad, S. Pediatric Liver Transplantation from a Living Donor in Mitochondrial Disease: Good Outcomes in DGUOK Deficiency? Pediatr. Transpl. 2020, 24, e13714. [Google Scholar] [CrossRef]
  135. Tam, A.; AlDhaheri, N.S.; Mysore, K.; Tessier, M.E.; Goss, J.; Fernandez, L.A.; D’Alessandro, A.M.; Schwoerer, J.S.; Rice, G.M.; Elsea, S.H.; et al. Improved Clinical Outcome Following Liver Transplant in Patients with Ethylmalonic Encephalopathy. Am. J. Med. Genet. A 2019, 179, 1015–1019. [Google Scholar] [CrossRef] [PubMed]
  136. Chinnery, P.F.; Jones, S.; Sviland, L.; Andrews, R.M.; Parsons, T.J.; Turnbull, D.M.; Bindoff, L.A. Mitochondrial Enteropathy: The Primary Pathology May Not Be within the Gastrointestinal Tract. Gut 2001, 48, 121–124. [Google Scholar] [CrossRef] [PubMed]
  137. Filosto, M.; Piccinelli, S.C.; Caria, F.; Cassarino, S.G.; Baldelli, E.; Galvagni, A.; Volonghi, I.; Scarpelli, M.; Padovani, A. Mitochondrial Neurogastrointestinal Encephalomyopathy (MNGIE-MTDPS1). J. Clin. Med. 2018, 7, 389. [Google Scholar] [CrossRef] [PubMed]
  138. Hirano, M.; Carelli, V.; De Giorgio, R.; Pironi, L.; Accarino, A.; Cenacchi, G.; D’Alessandro, R.; Filosto, M.; Martí, R.; Nonino, F.; et al. Mitochondrial Neurogastrointestinal Encephalomyopathy (MNGIE): Position Paper on Diagnosis, Prognosis, and Treatment by the MNGIE International Network. J. Inherit. Metab. Dis. 2021, 44, 376–387. [Google Scholar] [CrossRef] [PubMed]
  139. Ng, Y.S.; Feeney, C.; Schaefer, A.M.; Holmes, C.E.; Hynd, P.; Alston, C.L.; Grady, J.P.; Roberts, M.; Maguire, M.; Bright, A.; et al. Pseudo-Obstruction, Stroke, and Mitochondrial Dysfunction: A Lethal Combination. Ann. Neurol. 2016, 80, 686–692. [Google Scholar] [CrossRef]
  140. Hikmat, O.; Charalampos, T.; Klingenberg, C.; Rasmussen, M.; Tallaksen, C.M.E.; Brodtkorb, E.; Fiskerstrand, T.; McFarland, R.; Rahman, S.; Bindoff, L.A. The Presence of Anaemia Negatively Influences Survival in Patients with POLG Disease. J. Inherit. Metab. Dis. 2017, 40, 861–866. [Google Scholar] [CrossRef]
  141. Prasun, P.; Koeberl, D.D. Mitochondrial Neurogastrointestinal Encephalomyopathy (MNGIE)-like Phenotype in a Patient with a Novel Heterozygous POLG Mutation. J. Neurol. 2014, 261, 1818–1819. [Google Scholar] [CrossRef]
  142. Tang, S.; Dimberg, E.L.; Milone, M.; Wong, L.J.C. Mitochondrial Neurogastrointestinal Encephalomyopathy (MNGIE)-like Phenotype: An Expanded Clinical Spectrum of POLG1 Mutations. J. Neurol. 2012, 259, 862–868. [Google Scholar] [CrossRef]
  143. El-Hattab, A.W.; Wang, J.; Dai, H.; Almannai, M.; Scaglia, F.; Craigen, W.J.; Wong, L.-J.C. MPV17-Related Mitochondrial DNA Maintenance Defect. In GeneReviews®; University of Washington, Seattle: Seattle, WA, USA, 2012. [Google Scholar]
  144. Scaglia, F.; Towbin, J.A.; Craigen, W.J.; Belmont, J.W.; Smith, E.O.B.; Neish, S.R.; Ware, S.M.; Hunter, J.V.; Fernbach, S.D.; Vladutiu, G.D.; et al. Clinical Spectrum, Morbidity, and Mortality in 113 Pediatric Patients with Mitochondrial Disease. Pediatrics 2004, 114, 925–931. [Google Scholar] [CrossRef]
  145. Holmgren, D.; Wåhlander, H.; Eriksson, B.O.; Oldfors, A.; Holme, E.; Tulinius, M. Cardiomyopathy in Children with Mitochondrial Disease: Clinical Course and Cardiological Findings. Eur. Heart J. 2003, 24, 280–288. [Google Scholar] [CrossRef]
  146. Sofou, K.; De Coo, I.F.M.; Isohanni, P.; Ostergaard, E.; Naess, K.; De Meirleir, L.; Tzoulis, C.; Uusimaa, J.; De Angst, I.B.; Lönnqvist, T.; et al. A Multicenter Study on Leigh Syndrome: Disease Course and Predictors of Survival. Orphanet. J. Rare Dis. 2014, 9, 52. [Google Scholar] [CrossRef] [PubMed]
  147. Clarke, S.L.; Bowron, A.; Gonzalez, I.L.; Groves, S.J.; Newbury-Ecob, R.; Clayton, N.; Martin, R.P.; Tsai-Goodman, B.; Garratt, V.; Ashworth, M.; et al. Barth Syndrome. Orphanet. J. Rare Dis. 2013, 8, 23. [Google Scholar] [CrossRef] [PubMed]
  148. Suzuki-Hatano, S.; Saha, M.; Rizzo, S.A.; Witko, R.L.; Gosiker, B.J.; Ramanathan, M.; Soustek, M.S.; Jones, M.D.; Kang, P.B.; Byrne, B.J.; et al. AAV-Mediated TAZ Gene Replacement Restores Mitochondrial and Cardioskeletal Function in Barth Syndrome. Hum. Gene Ther. 2019, 30, 139–154. [Google Scholar] [CrossRef]
  149. Kang, Y.; Stroud, D.A.; Baker, M.J.; De Souza, D.P.; Frazier, A.E.; Liem, M.; Tull, D.; Mathivanan, S.; McConville, M.J.; Thorburn, D.R.; et al. Sengers Syndrome-Associated Mitochondrial Acylglycerol Kinase Is a Subunit of the Human TIM22 Protein Import Complex. Mol. Cell 2017, 67, 457–470.e5. [Google Scholar] [CrossRef] [PubMed]
  150. Vukotic, M.; Nolte, H.; König, T.; Saita, S.; Ananjew, M.; Krüger, M.; Tatsuta, T.; Langer, T. Acylglycerol Kinase Mutated in Sengers Syndrome Is a Subunit of the TIM22 Protein Translocase in Mitochondria. Mol. Cell 2017, 67, 471–483.e7. [Google Scholar] [CrossRef] [PubMed]
  151. Enns, G.M. Pediatric Mitochondrial Diseases and the Heart. Curr. Opin. Pediatr. 2017, 29, 541–551. [Google Scholar] [CrossRef] [PubMed]
  152. Finsterer, J.; Kothari, S. Cardiac Manifestations of Primary Mitochondrial Disorders. Int. J. Cardiol. 2014, 177, 754–763. [Google Scholar] [CrossRef]
  153. Akaike, M.; Kawai, H.; Yokoi, K.; Kunishige, M.; Mine, H.; Nishida, Y.; Saito, S. Cardiac Dysfunction in Patients with Chronic Progressive External Ophthalmoplegia. Clin. Cardiol. 1997, 20, 239–243. [Google Scholar] [CrossRef]
  154. Kennedy, H.; Haack, T.B.B.; Hartill, V.; Mataković, L.; Baumgartner, E.R.; Potter, H.; Mackay, R.; Alston, C.L.L.; O’Sullivan, S.; McFarland, R.; et al. Sudden Cardiac Death Due to Deficiency of the Mitochondrial Inorganic Pyrophosphatase PPA2. Am. J. Hum. Genet. 2016, 99, 674–682. [Google Scholar] [CrossRef]
  155. Quadir, A.; Pontifex, C.S.; Robertson, H.L.; Labos, C.; Pfeffer, G. Systematic Review and Meta-Analysis of Cardiac Involvement in Mitochondrial Myopathy. Neurol. Genet. 2019, 5, e339. [Google Scholar] [CrossRef]
  156. Fang, W.; Huang, C.-C.; Chu, N.-S.; Lee, C.-C.; Chen, R.-S.; Pang, C.-Y.; Shih, K.-D.; Wei, Y.-H. Myoclonic Epilepsy with Ragged-Red Fibers (MERRF) Syndrome: Report of a Chinese Family with Mitochondrial DNA Point Mutation in TRNALys Gene. Muscle Nerve 1994, 17, 52–57. [Google Scholar] [CrossRef]
  157. Lorenzoni, P.J.; Scola, R.H.; Kay, C.S.K.; Arndt, R.C.; Silvado, C.E.; Werneck, L.C. MERRF: Clinical Features, Muscle Biopsy and Molecular Genetics in Brazilian Patients. Mitochondrion 2011, 11, 528–532. [Google Scholar] [CrossRef]
  158. Catteruccia, M.; Sauchelli, D.; Della Marca, G.; Primiano, G.; Cuccagna, C.; Bernardo, D.; Leo, M.; Camporeale, A.; Sanna, T.; Cianfoni, A.; et al. “Myo-Cardiomyopathy” Is Commonly Associated with the A8344G “MERRF” Mutation. J. Neurol. 2015, 262, 701–710. [Google Scholar] [CrossRef] [PubMed]
  159. Santorelli, F.M.; Mak, S.C.; El-Schahawi, M.; Casali, C.; Shanske, S.; Baram, T.Z.; Madrid, R.E.; DiMauro, S. Maternally Inherited Cardiomyopathy and Hearing Loss Associated with a Novel Mutation in the Mitochondrial TRNA(Lys) Gene (G8363A). Am. J. Hum. Genet. 1996, 58, 933–939. [Google Scholar]
  160. Mollet, J.; Giurgea, I.; Schlemmer, D.; Dallner, G.; Chretien, D.; Delahodde, A.; Bacq, D.; De Lonlay, P.; Munnich, A.; Rötig, A. Prenyldiphosphate Synthase, Subunit 1 (PDSS1) and OH-Benzoate Polyprenyltransferase (COQ2) Mutations in Ubiquinone Deficiency and Oxidative Phosphorylation Disorders. J. Clin. Investig. 2007, 117, 765–772. [Google Scholar] [CrossRef] [PubMed]
  161. Crow, Y.; Keshavan, N.; Barbet, J.P.; Bercu, G.; Bondet, V.; Boussard, C.; Dedieu, N.; Duffy, D.; Hully, M.; Giardini, A.; et al. Cardiac Valve Involvement in ADAR-Related Type I Interferonopathy. J. Med. Genet. 2020, 57, 475–478. [Google Scholar] [CrossRef]
  162. Bagnasco, S.; Good, D.; Balaban, R.; Burg, M. Lactate Production in Isolated Segments of the Rat Nephron. Am. J. Physiol. 1985, 248, F522–F526. [Google Scholar] [CrossRef] [PubMed]
  163. Lee, Y.S.; Yap, H.K.; Barshop, B.A.; Lee, Y.S.; Rajalingam, S.; Loke, K.Y. Mitochondrial Tubulopathy: The Many Faces of Mitochondrial Disorders. Pediatr. Nephrol. 2001, 16, 710–712. [Google Scholar] [CrossRef]
  164. Martín-Hernández, E.; García-Silva, M.T.; Vara, J.; Campos, Y.; Cabello, A.; Muley, R.; del Hoyo, P.; Martín, M.A.; Arenas, J. Renal Pathology in Children with Mitochondrial Diseases. Pediatr. Nephrol. 2005, 20, 1299–1305. [Google Scholar] [CrossRef]
  165. Rahman, S.; Hall, A.M. Mitochondrial Disease—An Important Cause of End-Stage Renal Failure. Pediatr. Nephrol. 2012, 28, 357–361. [Google Scholar] [CrossRef]
  166. Emma, F.; Bertini, E.; Salviati, L.; Montini, G. Renal Involvement in Mitochondrial Cytopathies. Pediatr. Nephrol. 2012, 27, 539–550. [Google Scholar] [CrossRef] [PubMed]
  167. Hall, A.M.; Vilasi, A.; Garcia-Perez, I.; Lapsley, M.; Alston, C.L.; Pitceathly, R.D.S.; McFarland, R.; Schaefer, A.M.; Turnbull, D.M.; Beaumont, N.J.; et al. The Urinary Proteome and Metabonome Differ from Normal in Adults with Mitochondrial Disease. Kidney Int. 2015, 87, 610–622. [Google Scholar] [CrossRef] [PubMed]
  168. Shayota, B.J.; Le, N.T.; Bekheirnia, N.; Rosenfeld, J.A.; Goldstein, A.C.; Moritz, M.; Bartholomew, D.W.; Pastore, M.T.; Xia, F.; Eng, C.; et al. Characterization of the Renal Phenotype in RMND1-Related Mitochondrial Disease. Mol. Genet. Genom. Med. 2019, 7, e973. [Google Scholar] [CrossRef] [PubMed]
  169. Ozaltin, F. Primary Coenzyme Q10 (CoQ10) Deficiencies and Related Nephropathies. Pediatr. Nephrol. 2014, 29, 961–969. [Google Scholar] [CrossRef]
  170. Chow, J.; Rahman, J.; Achermann, J.C.; Dattani, M.T.; Rahman, S. Mitochondrial Disease and Endocrine Dysfunction. Nat. Rev. Endocrinol. 2017, 13, 92–104. [Google Scholar] [CrossRef] [PubMed]
  171. Maechler, P. Mitochondrial Function and Insulin Secretion. Mol. Cell. Endocrinol. 2013, 379, 12–18. [Google Scholar] [CrossRef]
  172. Karaa, A.; Goldstein, A. The Spectrum of Clinical Presentation, Diagnosis, and Management of Mitochondrial Forms of Diabetes. Pediatr. Diabetes 2015, 16, 1–9. [Google Scholar] [CrossRef]
  173. Pagnamenta, A.T.; Taanman, J.W.; Wilson, C.J.; Anderson, N.E.; Marotta, R.; Duncan, A.J.; Bitner-Glindzicz, M.; Taylor, R.W.; Laskowski, A.; Thorburn, D.R.; et al. Dominant Inheritance of Premature Ovarian Failure Associated with Mutant Mitochondrial DNA Polymerase Gamma. Hum. Reprod. 2006, 21, 2467–2473. [Google Scholar] [CrossRef] [PubMed]
  174. Dallabona, C.; Diodato, D.; Kevelam, S.H.; Haack, T.B.; Wong, L.-J.; Salomons, G.S.; Baruffini, E.; Melchionda, L.; Mariotti, C.; Strom, T.M.; et al. Novel (Ovario) Leukodystrophy Related to AARS2 Mutations. Neurology 2014, 82, 2063–2071. [Google Scholar] [CrossRef]
  175. Fassone, E.; Rahman, S. Complex I Deficiency: Clinical Features, Biochemistry and Molecular Genetics. J. Med. Genet. 2012, 49, 578–590. [Google Scholar] [CrossRef]
  176. Glamuzina, E.; Brown, R.; Hogarth, K.; Saunders, D.; Russell-Eggitt, I.; Pitt, M.; De Sousa, C.; Rahman, S.; Brown, G.; Grunewald, S. Further Delineation of Pontocerebellar Hypoplasia Type 6 Due to Mutations in the Gene Encoding Mitochondrial Arginyl-TRNA Synthetase, RARS2. J. Inherit. Metab. Dis. 2012, 35, 459–467. [Google Scholar] [CrossRef]
  177. Jackson, M.J.; Schaefer, J.A.; Johnson, M.A.; Morris, A.A.M.; Turnbull, D.M.; Bindoff, L.A. Presentation and Clinical Investigation of Mitochondrial Respiratory Chain Disease A Study of 51 Patients. Brain 1995, 118, 339–357. [Google Scholar] [CrossRef]
  178. Munnich, A.; Rotig, A.; Chretien, D.; Cormier, V.; Bourgeron, T.; Bonnefont, J.-R.; Saudubray, J.-M.; Rustin, P. Clinical Presentation of Mitochondrial Disorders in Childhood. Inherit. Metab. Dis. 1996, 19, 521–527. [Google Scholar] [CrossRef]
  179. Lamont, P.J.; Surtees, R.; Woodward, C.E.; Leonard, J.V.; Wood, N.W.; Harding, A.E. Clinical and Laboratory Findings in Referrals for Mitochondrial DNA Analysis. Arch. Dis. Child. 1998, 79, 22–27. [Google Scholar] [CrossRef] [PubMed]
  180. Hutchesson, A.; Anne Preece, M.; Gray, G.; Green, A. Measurement of Lactate in Cerebrospinal Fluid in Investigation of Inherited Metabolic Disease. Clin. Chem. 1997, 43, 158–161. [Google Scholar] [CrossRef] [PubMed]
  181. Poggi-Travert, F.; Martin, D.; Billette De Villemeur, T.; Bonnefont, J.P.; Vassault, A.; Rabier, D.; Charpentier, C.; Kamoun, P.; Munnich, A.; Saudubray, J.M. Metabolic Intermediates in Lactic Acidosis: Compounds, Samples and Interpretation. J. Inherit. Metab. Dis. 1996, 19, 478–488. [Google Scholar] [CrossRef] [PubMed]
  182. Argilés, J.M.; Busquets, S.; Stemmler, B.; López-Soriano, F.J. Cancer Cachexia: Understanding the Molecular Basis. Nat. Rev. Cancer. 2014, 14, 754–762. [Google Scholar] [CrossRef] [PubMed]
  183. Fontes-Oliveira, C.C.; Busquets, S.; Toledo, M.; Penna, F.; Aylwin, M.P.; Sirisi, S.; Silva, A.P.; Orpí, M.; García, A.; Sette, A.; et al. Mitochondrial and Sarcoplasmic Reticulum Abnormalities in Cancer Cachexia: Altered Energetic Efficiency? Biochim. Biophys. Acta Gen. Subj. 2013, 1830, 2770–2778. [Google Scholar] [CrossRef]
  184. Shum, A.M.Y.; Mahendradatta, T.; Taylor, R.J.; Painter, A.B.; Moore, M.M.; Tsoli, M.; Tan, T.C.; Clarke, S.J.; Robertson, G.R.; Polly, P. Disruption of MEF2C Signaling and Loss of Sarcomeric and Mitochondrial Integrity in Cancer-Induced Skeletal Muscle Wasting. Aging 2012, 4, 133–143. [Google Scholar] [CrossRef] [PubMed]
  185. Brown, J.L.; Rosa-Caldwell, M.E.; Lee, D.E.; Blackwell, T.A.; Brown, L.A.; Perry, R.A.; Haynie, W.S.; Hardee, J.P.; Carson, J.A.; Wiggs, M.P.; et al. Mitochondrial Degeneration Precedes the Development of Muscle Atrophy in Progression of Cancer Cachexia in Tumour-Bearing Mice. J. Cachexia Sarcopenia Muscle 2017, 8, 926–938. [Google Scholar] [CrossRef]
  186. Porporato, P.E. Understanding Cachexia as a Cancer Metabolism Syndrome. Oncogenesis 2016, 5, e200. [Google Scholar] [CrossRef] [PubMed]
  187. Aversa, Z.; Pin, F.; Lucia, S.; Penna, F.; Verzaro, R.; Fazi, M.; Colasante, G.; Tirone, A.; Fanelli, F.R.; Ramaccini, C.; et al. Autophagy Is Induced in the Skeletal Muscle of Cachectic Cancer Patients. Sci. Rep. 2016, 6, 30340. [Google Scholar] [CrossRef]
  188. Busquets, S.; Deans, C.; Figueras, M.; Moore-Carrasco, R.; López-Soriano, F.J.; Fearon, K.C.H.; Argilés, J.M. Apoptosis Is Present in Skeletal Muscle of Cachectic Gastro-Intestinal Cancer Patients. Clin. Nutr. 2007, 26, 614–618. [Google Scholar] [CrossRef]
  189. Musumeci, O.; Barca, E.; Lamperti, C.; Servidei, S.; Comi, G.P.; Moggio, M.; Mongini, T.; Siciliano, G.; Filosto, M.; Pegoraro, E.; et al. Lipomatosis Incidence and Characteristics in an Italian Cohort of Mitochondrial Patients. Front. Neurol. 2019, 10, 160. [Google Scholar] [CrossRef]
  190. Klopstock, T.; Naumann, M.; Schalke, B.; Bischof, F.; Seibel, P.; Kottlors, M.; Eckert, P.; Reiners, K.; Toyka, K.V.; Reichmann, H. Multiple Symmetric Lipomatosis: Abnormalities in Complex IV and Multiple Deletions in Mitochondrial DNA. Neurology 1994, 44, 862. [Google Scholar] [CrossRef]
  191. Naumann, M.; Kiefer, R.; Toyka, K.V.; Sommer, C.; Seibel, P.; Reichmann, H. Mitochondrial Dysfunction with Myoclonus Epilepsy and Ragged Red Fibers Point Mutation in Nerve, Muscle, and Adipose Tissue of a Patient with Multiple Symmetric Lipomatosis. Muscle Nerve 1997, 20, 833–839. [Google Scholar] [CrossRef]
  192. Coin, A.; Enzi, G.; Bussolotto, M.; Ceschin, E.; Difito, M.; Angelini, C. Multiple Symmetric Lipomatosis. J. Clin. Neuromuscul. Dis. 2000, 1, 124–130. [Google Scholar] [CrossRef]
  193. Sawyer, S.L.; Ng, A.C.H.; Micheil Innes, A.; Wagner, J.D.; Dyment, D.A.; Tetreault, M.; Majewski, J.; Boycott, K.M.; Screaton, R.A.; Nicholson, G.; et al. Homozygous Mutations in MFN2 Cause Multiple Symmetric Lipomatosis Associated with Neuropathy. Hum. Mol. Genet. 2015, 24, 5109–5114. [Google Scholar] [CrossRef]
  194. Wolny, S.; McFarland, R.; Chinnery, P.; Cheetham, T. Abnormal Growth in Mitochondrial Disease. Acta Paediatr. Int. J. Paediatr. 2009, 98, 553–554. [Google Scholar] [CrossRef] [PubMed]
  195. Castro-Gago, M.; Gómez-Lado, C.; Pérez-Gay, L.; Eirís-Puñal, J. Abnormal Growth in Mitochondrial Disease. Acta Paediatr. Int. J. Paediatr. 2010, 99, 796. [Google Scholar] [CrossRef]
  196. Xia, C.Y.; Liu, Y.; Liu, H.; Zhang, Y.C.; Ma, Y.N.; Qi, Y. Clinical and Molecular Characteristics in 100 Chinese Pediatric Patients with m.3243a>G Mutation in Mitochondrial DNA. Chin. Med. J. 2016, 129, 1945–1949. [Google Scholar] [CrossRef]
  197. Koenig, M.K.; Grant, L. Neuropathy, Ataxia, and Retinitis Pigmentosa. In Mitochondrial Case Studies; Elsevier: Amsterdam, The Netherlands, 2016; pp. 69–73. [Google Scholar]
  198. Matsuzaki, M.; Izumi, T.; Shishikura, K.; Suzuki, H.; Hirayama, Y. Hypothalamic Growth Hormone Deficiency and Supplementary GH Therapy in Two Patients with Mitochondrial Myopathy, Encephalopathy, Lactic Acidosis and Stroke-like Episodes. Neuropediatrics 2002, 33, 271–273. [Google Scholar] [CrossRef]
  199. Broomfield, A.; Sweeney, M.G.; Woodward, C.E.; Fratter, C.; Morris, A.M.; Leonard, J.V.; Abulhoul, L.; Grunewald, S.; Clayton, P.T.; Hanna, M.G.; et al. Paediatric Single Mitochondrial DNA Deletion Disorders: An Overlapping Spectrum of Disease. J. Inherit. Metab. Dis. 2015, 38, 445–457. [Google Scholar] [CrossRef]
  200. Shiraiwa, N.; Ishii, A.; Iwamoto, H.; Mizusawa, H.; Kagawa, Y.; Ohta, S. Content of Mutant Mitochondrial DNA and Organ Dysfunction in a Patient with a MELAS Subgroup of Mitochondrial Encephalomyopathies. J. Neurol. Sci. 1993, 120, 174–179. [Google Scholar] [CrossRef]
  201. De Laat, P.; Zweers, H.E.; Knuijt, S.; Smeitink, J.A.; Wanten, G.J.; Janssen, M.C. Dysphagia, Malnutrition and Gastrointestinal Problems in Patients with Mitochondrial Disease Caused by the M3243A>G Mutation. Neth. J. Med. 2015, 73, 30–36. [Google Scholar]
  202. Nooteboom, M.; Johnson, R.; Taylor, R.W.; Wright, N.A.; Lightowlers, R.N.; Kirkwood, T.B.L.; Mathers, J.C.; Turnbull, D.M.; Greaves, L.C. Age-Associated Mitochondrial DNA Mutations Lead to Small but Significant Changes in Cell Proliferation and Apoptosis in Human Colonic Crypts. Aging Cell 2010, 9, 96–99. [Google Scholar] [CrossRef]
  203. Gandhi, S.S.; Muraresku, C.; McCormick, E.M.; Falk, M.J.; McCormack, S.E. Risk Factors for Poor Bone Health in Primary Mitochondrial Disease. J. Inherit. Metab. Dis. 2017, 40, 673–683. [Google Scholar] [CrossRef]
  204. Wong, S.C.; Dobie, R.; Altowati, M.A.; Werther, G.A.; Farquharson, C.; Ahmed, S.F. Growth and the Growth Hormone-Insulin like Growth Factor 1 Axis in Children with Chronic Inflammation: Current Evidence, Gaps in Knowledge, and Future Directions. Endocr. Rev. 2016, 37, 62–110. [Google Scholar] [CrossRef]
  205. Schaefer, A.M.; Walker, M.; Turnbull, D.M.; Taylor, R.W. Endocrine Disorders in Mitochondrial Disease. Mol. Cell Endocrinol. 2013, 379, 2–11. [Google Scholar] [CrossRef]
  206. Carelli, V.; Musumeci, O.; Caporali, L.; Zanna, C.; La Morgia, C.; Del Dotto, V.; Porcelli, A.M.; Rugolo, M.; Valentino, M.L.; Iommarini, L.; et al. Syndromic Parkinsonism and Dementia Associated with OPA1 Missense Mutations. Ann. Neurol. 2015, 78, 21–38. [Google Scholar] [CrossRef] [PubMed]
  207. Samocha-Bonet, D.; Wu, B.; Ryugo, D.K. Diabetes Mellitus and Hearing Loss: A Review. Ageing Res. Rev. 2021, 71, 101423. [Google Scholar] [CrossRef]
  208. Ali, A.S.; Ekinci, E.I.; Pyrlis, F. Maternally Inherited Diabetes and Deafness (MIDD): An Uncommon but Important Cause of Diabetes. Endocr. Metab. Sci. 2021, 2, 100074. [Google Scholar] [CrossRef]
  209. Urano, F. Wolfram Syndrome: Diagnosis, Management, and Treatment. Curr. Diab. Rep. 2016, 16, 6. [Google Scholar] [CrossRef]
  210. Sako, S.; Tsunogai, T.; Oishi, K. Thiamine-Responsive Megaloblastic Anemia Syndrome. In GeneReviews®; University of Washington, Seattle: Seattle, WA, USA, 2003. [Google Scholar]
  211. OMIM®—Online Mendelian Inheritance in Man®. Available online: http://www.omim.org/ (accessed on 26 July 2023).
  212. Alharbi, H.; Priestley, J.R.C.; Wilkins, B.J.; Ganetzky, R.D. Mitochondrial Hepatopathies. Clin. Liver. Dis. 2021, 18, 243–250. [Google Scholar] [CrossRef]
  213. Khurana, D.S.; Salganicoff, L.; Melvin, J.J.; Hobdell, E.F.; Valencia, I.; Hardison, H.H.; Marks, H.G.; Grover, W.D.; Legido, A. Epilepsy and Respiratory Chain Defects in Children with Mitochondrial Encephalopathies. Neuropediatrics 2008, 39, 8–13. [Google Scholar] [CrossRef]
  214. Bindoff, L.A.; Engelsen, B.A. Mitochondrial Diseases and Epilepsy. Epilepsia 2012, 53, 92–97. [Google Scholar] [CrossRef]
  215. Harris, S.R. Congenital Hypotonia: Clinical and Developmental Assessment. Dev. Med. Child. Neurol. 2008, 50, 889–892. [Google Scholar] [CrossRef]
  216. Pinto, M.; Pickrell, A.M.; Moraes, C.T. Regional Susceptibilities to Mitochondrial Dysfunctions in the CNS. Biol. Chem. 2012, 393, 275–281. [Google Scholar] [CrossRef]
  217. Turconi, A.C.; Benti, R.; Castelli, E.; Pochintesta, S.; Felisari, G.; Comi, G.; Gagliardi, C.; Del Piccolo, L.; Bresolin, N. Focal Cognitive Impairment in Mitochondrial Encephalomyopathies: A Neuropsychological and Neuroimaging Study. J. Neurol. Sci. 1999, 170, 57–63. [Google Scholar] [CrossRef] [PubMed]
  218. Palmieri, L.; Persico, A.M. Mitochondrial Dysfunction in Autism Spectrum Disorders: Cause or Effect? Bio-Chim. Biophys. Acta Bioenerg. 2010, 1797, 1130–1137. [Google Scholar] [CrossRef] [PubMed]
  219. DiMauro, S.; Schon, E.A. Mitochondrial Disorders in the Nervous System. Annu. Rev. Neurosci. 2008, 31, 91–123. [Google Scholar] [CrossRef] [PubMed]
  220. Parikh, S.; Goldstein, A.; Koenig, M.K.; Scaglia, F.; Enns, G.M.; Saneto, R.; Anselm, I.; Cohen, B.H.; Falk, M.J.; Greene, C.; et al. Diagnosis and Management of Mitochondrial Disease: A Consensus Statement from the Mitochondrial Medicine Society. Genet. Med. 2015, 17, 689–701. [Google Scholar] [CrossRef] [PubMed]
  221. Mavraki, E.; Labrum, R.; Sergeant, K.; Alston, C.L.; Woodward, C.; Smith, C.; Knowles, C.V.Y.; Patel, Y.; Hodsdon, P.; Baines, J.P.; et al. Genetic Testing for Mitochondrial Disease: The United Kingdom Best Practice Guidelines. Eur. J. Hum. Genet. 2023, 31, 148–163. [Google Scholar] [CrossRef]
  222. Almannai, M.; El-Hattab, A.W.; Ali, M.; Soler-Alfonso, C.; Scaglia, F. Clinical Trials in Mitochondrial Disorders, an Update. Mol. Genet. Metab. 2020, 131, 1–13. [Google Scholar] [CrossRef]
  223. Lynch, D.R.; Chin, M.P.; Boesch, S.; Delatycki, M.B.; Giunti, P.; Goldsberry, A.; Hoyle, J.C.; Mariotti, C.; Mathews, K.D.; Nachbauer, W.; et al. Efficacy of Omaveloxolone in Friedreich’s Ataxia: Delayed-Start Analysis of the MOXIe Extension. Mov. Disord. 2023, 38, 313–320. [Google Scholar] [CrossRef] [PubMed]
  224. Domínguez-González, C.; Madruga-Garrido, M.; Mavillard, F.; Garone, C.; Aguirre-Rodríguez, F.J.; Donati, M.A.; Kleinsteuber, K.; Martí, I.; Martín-Hernández, E.; Morealejo-Aycinena, J.P.; et al. Deoxynucleoside Therapy for Thymidine Kinase 2-Deficient Myopathy. Ann. Neurol. 2019, 86, 293–303. [Google Scholar] [CrossRef] [PubMed]
  225. Yu-Wai-Man, P.; Newman, N.J.; Carelli, V.; Moster, M.L.; Biousse, V.; Sadun, A.A.; Klopstock, T.; Vignal-Clermont, C.; Sergott, R.C.; Rudolph, G.; et al. Bilateral Visual Improvement with Unilateral Gene Therapy Injection for Leber Hereditary Optic Neuropathy. Sci. Transl. Med. 2020, 12, eaaz7423. [Google Scholar] [CrossRef]
  226. Soldatov, V.O.; Kubekina, M.V.; Skorkina, M.Y.; Belykh, A.E.; Egorova, T.V.; Korokin, M.V.; Pokrovskiy, M.V.; Deykin, A.V.; Angelova, P.R. Current Advances in Gene Therapy of Mitochondrial Diseases. J. Transl. Med. 2022, 20, 562. [Google Scholar] [CrossRef]
  227. Yamada, K.; Toribe, Y.; Yanagihara, K.; Mano, T.; Akagi, M.; Suzuki, Y. Diagnostic Accuracy of Blood and CSF Lactate in Identifying Children with Mitochondrial Diseases Affecting the Central Nervous System. Brain Dev. 2012, 34, 92–97. [Google Scholar] [CrossRef]
  228. Yatsuga, S.; Fujita, Y.; Ishii, A.; Fukumoto, Y.; Arahata, H.; Kakuma, T.; Kojima, T.; Ito, M.; Tanaka, M.; Saiki, R.; et al. Growth Differentiation Factor 15 as a Useful Biomarker for Mitochondrial Disorders. Ann. Neurol. 2015, 78, 814–823. [Google Scholar] [CrossRef]
  229. Mancuso, M.; Orsucci, D.; Coppedè, F.; Nesti, C.; Choub, A.; Siciliano, G. Diagnostic approach to mitochondrial disorders: The need for a reliable biomarker. Curr. Mol. Med. 2009, 9, 1095–1107. [Google Scholar] [CrossRef] [PubMed]
  230. Suomalainen, A.; Elo, J.M.; Pietiläinen, K.H.; Hakonen, A.H.; Sevastianova, K.; Korpela, M.; Isohanni, P.; Marjavaara, S.K.; Tyni, T.; Kiuru-Enari, S.; et al. FGF-21 as a Biomarker for Muscle-Manifesting Mitochondrial Respiratory Chain Deficiencies: A Diagnostic Study. Lancet Neurol. 2011, 10, 806–818. [Google Scholar] [CrossRef] [PubMed]
  231. Suomalainen, A. Fibroblast Growth Factor 21: A Novel Biomarker for Human Muscle-Manifesting Mitochondrial Disorders. Expert Opin. Med. Diagn. 2013, 7, 313–317. [Google Scholar] [CrossRef]
  232. Boenzi, S.; Diodato, D. Biomarkers for Mitochondrial Energy Metabolism Diseases. Essays Biochem. 2018, 62, 443–454. [Google Scholar] [CrossRef] [PubMed]
  233. Pajares, S.; Arias, A.; García-Villoria, J.; Briones, P.; Ribes, A. Role of Creatine as Biomarker of Mitochondrial Diseases. Mol. Genet. Metab. 2013, 108, 119–124. [Google Scholar] [CrossRef] [PubMed]
  234. Mitochondrial Medicine Society’s Committee on Diagnosis; Haas, R.H.; Parikh, S.; Falk, M.J.; Saneto, R.P.; Wolf, N.I.; Darin, N.; Wong, L.-J.; Cohen, B.H.; Naviaux, R.K. The In-Depth Evaluation of Suspected Mitochondrial Disease. Mol. Genet. Metab. 2008, 94, 16–37. [Google Scholar] [CrossRef] [PubMed]
  235. Ji, X.; Zhao, L.; Ji, K.; Zhao, Y.; Li, W.; Zhang, R.; Hou, Y.; Lu, J.; Yan, C. Growth Differentiation Factor 15 Is a Novel Diagnostic Biomarker of Mitochondrial Diseases. Mol. Neurobiol. 2017, 54, 8110–8116. [Google Scholar] [CrossRef]
  236. Lehtonen, J.M.; Forsström, S.; Bottani, E.; Viscomi, C.; Baris, O.R.; Isoniemi, H.; Höckerstedt, K.; Österlund, P.; Hurme, M.; Jylhävä, J.; et al. FGF21 Is a Biomarker for Mitochondrial Translation and MtDNA Maintenance Disorders. Neurology 2016, 87, 2290–2299. [Google Scholar] [CrossRef]
  237. Davis, R.L.; Liang, C.; Sue, C.M. A Comparison of Current Serum Biomarkers as Diagnostic Indicators of Mitochondrial Diseases. Neurology 2016, 86, 2010–2015. [Google Scholar] [CrossRef]
  238. Kalko, S.G.; Paco, S.; Jou, C.; Rodríguez, M.A.; Meznaric, M.; Rogac, M.; Jekovec-Vrhovsek, M.; Sciacco, M.; Moggio, M.; Fagiolari, G.; et al. Transcriptomic Profiling of TK2 Deficient Human Skeletal Muscle Suggests a Role for the P53 Signalling Pathway and Identifies Growth and Differentiation Factor-15 as a Potential Novel Biomarker for Mitochondrial Myopathies. BMC Genom. 2014, 15, 91. [Google Scholar] [CrossRef]
  239. Morovat, A.; Weerasinghe, G.; Nesbitt, V.; Hofer, M.; Agnew, T.; Quaghebeur, G.; Sergeant, K.; Fratter, C.; Guha, N.; Mirzazadeh, M.; et al. Use of FGF-21 as a Biomarker of Mitochondrial Disease in Clinical Practice. J. Clin. Med. 2017, 6, 80. [Google Scholar] [CrossRef]
  240. Baldacci, J.; Calderisi, M.; Fiorillo, C.; Santorelli, F.M.; Rubegni, A. Automatic Recognition of Ragged Red Fibers in Muscle Biopsy from Patients with Mitochondrial Disorders. Healthcare 2022, 10, 574. [Google Scholar] [CrossRef]
  241. Pfeffer, G.; Chinnery, P.F. Diagnosis and Treatment of Mitochondrial Myopathies. Ann. Med. 2013, 45, 4–16. [Google Scholar] [CrossRef]
Figure 1. Graphical illustration of the main mitochondrial manifestations and red flags divided by system/organs.
Figure 1. Graphical illustration of the main mitochondrial manifestations and red flags divided by system/organs.
Ijms 24 16746 g001
Table 1. Key phenotypic features, red flags, and main genetic etiology of some examples of PMDs discussed in this review.
Table 1. Key phenotypic features, red flags, and main genetic etiology of some examples of PMDs discussed in this review.
PMDKey Phenotype FeaturesMost Common Genotype
Well-defined syndromes
cPEOBilateral ptosis, progressive external
ophthalmoplegia, mild exercise intolerance
mtDNA single large-scale deletion, nuclear genes involved in mtDNA maintenance (e.g., POLG1) (autosomal cPEO associated with multiple mtDNA deletions)
KSSPEO (onset age < 20 years), pigmentary retinopathy, CSF protein >1 g/L, cerebellar ataxia, heart blockmtDNA single large-scale deletion
Pearson
syndrome
Sideroblastic anemia of childhood, pancytopenia, exocrine pancreatic failuremtDNA single large-scale deletion
MELASSLEs, epilepsy, lactic acidosis, dementia, diabetes mellitus, hearing loss, cardiomyopathym.3243A>G (MT-TL1)
Leigh
syndrome
Infantile onset, bilateral brainstem lesions with subacute relapsing encephalopathy, cerebellar and brain stem signsMT-ATP6
>100 nuclear-encoded genes (e.g., SURF1)
NARPPeripheral neuropathy, sensitive ataxia, pigmentary retinopathyMT-ATP6
MERRFMyoclonus, epilepsy, cerebellar ataxia, myopathym.8344A>G (MT-TK)
LHONSubacute painless bilateral visual failurem.11778G>A (MT-ND4), m.3460G>A (MT-ND1), m.14484T>C (MT-ND6)
ADOASubacute painless bilateral visual failureOPA1
MIDDHearing loss, diabetes mellitusm.3243A>G (MT-TL1)
MSLMultiple lipomas, myopathym.8344A>G (MT-TK), MFN2
PMMProximal myopathy with or without PEO, exercise intolerancemtDNA tRNAs (e.g., m.3243A>G in MT-TL1), mtDNA single large-scale deletion, POLG1, TWNK
Specific groups of disorders associated with multiple mtDNA deletions or mtDNA depletion
ANSMIRAS: mitochondrial recessive ataxia syndrome
SANDO: sensory ataxia, neuropathy, dysarthria, ophthalmoplegia
POLG1
MEMSAEpilepsy, myopathy, and ataxia without ophthalmoplegiaPOLG1
AHSEncephalopathy with intractable epilepsy, neuropathy, hepatic failurePOLG1
MCHSDevelopmental delay or dementia, lactic acidosis, myopathy with failure to thrive, hepatic failurePOLG1
MNGIESevere gastrointestinal dysmotility, cachexia, PEO,
sensorimotor neuropathy
TYMP
MDSMyopathic forms: hypotonia, myopathy, feeding difficulty (“floppy infant”)
Encephalomyopathic forms: hypotonia, global developmental delay
Hepatocerebral forms: hepatic failure, developmental delay, abnormal eye movements, peripheral neuropathy (e.g., MCHS, POLG-related)
Neurogastrointestinal forms: MNGIE
Mitochondrial nucleotide synthesis (TK2, SUCLA2, SUCLG1, RRM2B, DGUOK, TYMP, MPV17)
mtDNA replication (POLG1, TWNK)
cPEO: chronic progressive external pphthalmoplegia; KSS: Kearns–Sayre syndrome; CSF: cerebrospinal fluid; MELAS: mitochondrial encephalomyopathy with lactic acidosis and stroke-like episodes; SLEs: stroke-like episodes; NARP: neurogenic weakness with ataxia and retinitis pigmentosa; MERRF: myoclonic epilepsy with ragged-red fibers; LHON: Leber hereditary optic neuropathy; ADOA: autosomal dominant optic atrophy; MIDD: mitochondrial inherited diabetes and deafness; MSL: multiple systemic lipomatosis; PMM: primary mitochondrial myopathy; ANS: ataxia neuropathy spectrum; AHS: Alpers–Huttenlocher syndrome; MCHS: Childhood myocerebrohepatopathy spectrum; MNGIE: mitochondrial neurogastrointestinal encephalomyopathy; MDS: mitochondrial DNA depletion syndromes. Please note that MEMSA includes the syndrome previously described as spinocerebellar ataxia with epilepsy (SCAE).
Table 2. Primary mitochondrial diseases biomarkers.
Table 2. Primary mitochondrial diseases biomarkers.
BiomarkerDescriptionSamplesSP, SE, AUCReference
LactateQuantification of lactate, which results from anaerobic ATP productionBlood, CSF, urineSE: 34–62% SP: 83–100% [220]
AUC: 0.8–0.926 [227,228]
[229]
PyruvateQuantification of pyruvate, which is a precursor of lactateBlood, CSF, urineSE: 63.6–83.3%
SP: 87.2% [230]
AUC: 0.907 [227]
L/P ratio
SP: 100%
SE: 44% [231]
[232]
Creatine kinaseQuantification of creatine kinase, an enzyme released from damaged muscle fibersBlood, CSF, urineAUC: 0.609 [228][233]
AcylcarnitineQuantification of acylcarnitine, which transports activated long-chain fatty acids to mitochondria, required for beta oxidationBlood-[220]
Amino acidsQuantification of amino acids, as well as alanine, glycine, proline, and threonine, synthetized following respiratory chain dysfunction and altered cellular redox stateBlood, CSF, urine-[234]
GDF-15Quantification of GDF-15, a member of the transforming growth factor beta family, induced in response to cellular stressBloodAUC: 0.70–0.999 [235,236] SE: 77.8% [237][238]
FGF-21Quantification of FGF-21, a growth factor which regulates lipid and glucose metabolism, correlated with mitochondrial dysfunctionBloodAUC: 0.69–0.9 [230,239] SE: 41.7–95.7%
SP: 91.7% [230]
[239]
RRFsHistological identification of RRFs, which represent the accumulation of abnormal mitochondria below the plasma membrane of the muscle fiber, linked with mitochondrial dysfunction (modified Gomori trichrome staining)Muscle biopsy-[240]
RBFsHistological identification of RBFs, which represent muscle fibers with increased levels of succinate dehydrogenase activity in the subsarcolemmal regionMuscle biopsy-[241]
COX-negative fibersHistochemical identification of COX-deficient fibers, which represent a marker of mitochondrial respiratory chain deficiencyMuscle biopsy-[241]
SP: specificity; SE: sensitivity, AUC: area under curve; CSF: cerebrospinal fluid, RRFs: ragged red fibers, RBFs: ragged blue fibers, COX: cytochrome c oxidase. Note that the reported SP, SE, and AUC values refer to blood.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Conti, F.; Di Martino, S.; Drago, F.; Bucolo, C.; Micale, V.; Montano, V.; Siciliano, G.; Mancuso, M.; Lopriore, P. Red Flags in Primary Mitochondrial Diseases: What Should We Recognize? Int. J. Mol. Sci. 2023, 24, 16746. https://doi.org/10.3390/ijms242316746

AMA Style

Conti F, Di Martino S, Drago F, Bucolo C, Micale V, Montano V, Siciliano G, Mancuso M, Lopriore P. Red Flags in Primary Mitochondrial Diseases: What Should We Recognize? International Journal of Molecular Sciences. 2023; 24(23):16746. https://doi.org/10.3390/ijms242316746

Chicago/Turabian Style

Conti, Federica, Serena Di Martino, Filippo Drago, Claudio Bucolo, Vincenzo Micale, Vincenzo Montano, Gabriele Siciliano, Michelangelo Mancuso, and Piervito Lopriore. 2023. "Red Flags in Primary Mitochondrial Diseases: What Should We Recognize?" International Journal of Molecular Sciences 24, no. 23: 16746. https://doi.org/10.3390/ijms242316746

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop