Abstract Background The ATP synthase F1 subunit α (ATP5F1A) gene encodes a critical structural subunit of mitochondrial complex V. ATP5F1A mutations are linked to mitochondrial complex V deficiency diseases. Although only 14 cases have been reported globally, the genotype-phenotype correlations and underlying molecular mechanisms remain poorly understood. Objective To investigate the pathogenic mechanisms of ATP5F1A deficiency through functional analysis of a recurrent missense variant. Method A Han Chinese family with developmental delay and motor dysfunction was studied. Whole-exome sequencing and trio analysis identified the causative variant. Pathogenicity was evaluated using bioinformatic predictions and structural modeling. HEK293T cells were transfected with wild-type or mutant-type ATP5F1A plasmids for Western blot and immunofluorescence analysis. Morpholino (MO) oligonucleotides were microinjected into zebrafish embryos for gene knockdown. Motor neuron development was observed in Tg(mnx1:eGFP) zebrafish, with accompanying behavioral assessments. RNA sequencing was conducted to explore the underlying molecular pathways. Results A de novo missense variant (c.1252G > A, p.Gly418Arg) in ATP5F1A was identified and shown to segregate with the disease phenotype. The mutation reduced protein stability and expression. In HEK293T cells, the mutant protein exhibited reduced expression without affecting mitochondrial localization. In zebrafish, atp5fa1 knockdown caused growth retardation, motor dysfunction, and impaired motor neuron axon development. Rescue experiments with human wild-type ATP5F1A mRNA partially restored motor neuron morphology. Transcriptomic analysis identified 2,261 differentially expressed genes, enriched in neurotransmission and apelin signaling pathways. qPCR confirmed downregulation of autophagy-related genes (apln, becn1, map1lc3b) in knockdown larvae. Western blot showed that atp5fa1 knockdown increased P62 and decreased Lc3b-II expression in zebrafish models. Conclusion This study is the first to report pathogenic ATP5F1A mutations in the Chinese population. Atp5fa1 dysfunction leads to multi-system defects and disease phenotypes in a zebrafish model, possibly mediated through inhibiting autophagy activation mechanisms. Keywords: ATP5F1A, Complex v deficiency, Zebrafish, Neurodevelopmental deficits Introduction Primary mitochondrial diseases (PMDs) are inherited metabolic disorders caused by mutations in mitochondrial DNA (mtDNA) or nuclear DNA (nDNA) genes encoding components of the electron transport chain (ETC), with an incidence of approximately 1 in 5,000 [[32]1]. PMDs result from dysfunction in the mitochondrial oxidative phosphorylation system. As mitochondria are present in nearly all tissues and organs, clinical manifestations are highly heterogeneous and may affect multiple systems, including the nervous system, muscles, and heart [[33]2, [34]3]. Notably, childhood-onset PMDs are mainly due to nDNA mutations and are often characterized by early onset, severe symptoms, and high mortality [[35]4]. Mitochondrial complex V (ATP synthase) is the final effector of oxidative phosphorylation. Structural or functional abnormalities in this complex directly impair cellular energy metabolism. Complex V catalyzes the final step of oxidative phosphorylation during aerobic respiration by transferring protons across the mitochondrial electrochemical gradient and synthesizing ATP from ADP, inorganic phosphate (Pi), and Mg^2+ [[36]5]. It is encoded by two mtDNA genes—ATP6 and ATP8 (encoding subunits α and A6L, respectively)—and 27 nuclear genes. Its F1 head comprises three α and three β subunits arranged alternately to form the catalytic core [[37]6]. During ATP synthesis or hydrolysis, the α subunit provides essential arginine residues that assist the β subunit in catalysis, playing a key role in maintaining enzymatic activity [[38]7]. Mitochondrial complex V deficiency, a subtype of PMDs, is typically caused by mutations in structural subunits or assembly factors [[39]8]. Common clinical features include developmental delay, lactic acidosis, and hyperammonemia. Neurological symptoms may include epilepsy, various dystonias (including spasms and myoclonus), intellectual disability, and motor impairments [[40]9]. The ATP5F1A gene (OMIM#164360) located on chromosome 18q21.1, encodes the α subunit of the catalytic F1 domain of complex V, a core component of the mitochondrial ATP synthase complex [[41]10]. In 2013, ATP5F1A mutations were first reported to reduce complex V content and impair mitochondrial function, leading to fatal mitochondrial encephalopathy in infancy [[42]11]. Such mutations can disrupt ATP synthase activity, impair ATP production, and compromise cellular energy metabolism, resulting in various clinical symptoms [[43]12]. Currently, 14 cases linking ATP5F1A mutations to human diseases have been reported, with significant phenotypic heterogeneity depending on mutation sites and types. Recently, Harrer et al. [[44]13] reported the recurrent c.1252G > A variant in three individuals from two families with hereditary spastic paraplegia, providing the first clinical characterization of this mutation. Comprehensive molecular analysis, including fibroblast studies and structural modeling, confirmed its pathogenicity. However, the precise molecular mechanisms of ATP5F1A deficiency and its role in neurodevelopment remain unclear. In this study, we characterize the ATP5F1A c.1252G > A (p.Gly418Arg) variant, identified in a Han Chinese child with multi-system symptoms including growth retardation, movement disorders, and metabolic abnormalities. Through overexpression of the mutant gene and a zebrafish knockdown model combined with transcriptomic analysis, we found that ATP5F1A c.1252G > A is a loss-of-function (LoF) variant. The atp5fa1 MO zebrafish model replicated key phenotypes seen in patients. Disruption of the autophagy pathway may be a central mechanism in ATP5F1A-related neurodevelopmental disorders. Therefore, this study provides new insights into the genotype-phenotype correlations of ATP5F1A-associated diseases. Materials and methods Patient samples Blood samples were collected from the patients and their parents, and informed consent was obtained from the parents. The study was approved by the Ethical Review Committee of the Affiliated Hospital of Southwest Medical University (Ethics approval number: KY2025375). Molecular analysis Whole-exome sequencing of the family was used for the children and parents, followed by Sanger sequencing for validation. Briefly, DNA samples were extracted from peripheral blood, sequenced using high-throughput sequencing, and then filtered and screened for variants. Subsequently, primers were designed and Sanger sequencing was utilized to confirm the variant sites. Forward primer: 5′-GCTTGCTAGCGTGCGTATTT-3′; reverse primer: 5′-ACAACACGTAGTACAGGCCG-3′. In silico analysis For better clinical interpretation of variants, allele frequency data were obtained from the database gnomAD ([45]http://gnomad.broadinstitute.org/). We used the SIFT ([46]http://sift.jcvi.org), PolyPhen-2 ([47]http://genetics.bwh.harvard.edu/pph2) and MutationTaster ([48]http://www.mutationtaster.org) prediction tools to assess the pathogenicity of the variants and classified the variants according to the American College of Medical Genetics and Genomics (ACMG). We analyzed the amino acid conservation of the two mutation sites across species according to NCBI HomoloGene ([49]https://www.ncbi.nlm.nih.gov/homologene) and used the alignment function of MEGA ([50]https://www.megasoftware.net/) to compare multiple target sequences. Normal and mutant proteins were modeled using SwissModel ([51]http://swissmodel.expasy.org/) and visualized with the PyMOL package ([52]https://pymol.org/2/). Cell culture and transfection Human embryonic kidney 293T (HEK 293T) cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% fetal bovine serum (FBS) and 1% penicillin-streptomycin-amphotericin B at 37℃ with 5% CO[2]. The wild-type and c.1252G > A mutant full-length sequences encoding human ATP5F1A were cloned into the GV657 vector with a FLAG tag. For transfection experiments, HEK 293T cells (passages 3–7) were seeded in 6-well plates at a density of 3 × 10⁵ cells per well and cultured overnight until reaching a density of approximately 70%-80%. Transfection was performed using Lipofectamine 2000 (Invitrogen, Carlsbad, California, USA) according to the manufacturer’s instructions. Briefly, 2.5 µg of plasmid DNA was diluted in 200 µL of DMEM, and 5 µL of Lipofectamine 2000 was diluted in 200 µL of DMEM. After a 5-minute incubation at room temperature, the diluted DNA was mixed with Lipofectamine 2000 and incubated for an additional 20 min to form DNA-Lipofectamine complexes. The transfection mixture was then added dropwise to the cells cultured in antibiotic-free medium. Cells were collected for subsequent analysis 48–72 h post-transfection. qPCR At 72 h post-fertilization (hpf), zebrafish larvae were collected and homogenized for total RNA extraction using the TRNzol Universal Reagent (TianGen Biotech, Beijing, China), following the manufacturer’s instructions. First-strand cDNA was synthesized from 1 µg of total RNA using the PrimeScript™ RT Reagent Kit (Takara Bio Inc., Tokyo, Japan). Quantitative real-time PCR (RT-qPCR) was conducted using ChamQ Universal SYBR qPCR Master Mix (Vazyme Biotech, Nanjing, China) on a LightCycler^® 96 System (Roche Diagnostics, Basel, Switzerland). Each experimental group consisted of 15 zebrafish larvae, with three independent biological replicates and three technical replicates per sample. The relative mRNA expression levels were normalized to β-actin, which served as an internal reference gene, and were calculated using the 2^(-ΔΔCT) method. qPCR primers are shown in Table S1. Western blotting HEK 293T cells transfected with plasmids and zebrafish larvae at 72hpf were lysed in RIPA buffer (50 mM Tris-HCl pH 7.4, 150 mM NaCl, 1% Triton X-100, 1% sodium deoxycholate, 0.1% SDS; Beyotime, Shanghai, China) supplemented with protease inhibitor cocktail (Beyotime). Following centrifugation at 12,000 × g for 10 min at 4 °C, protein concentrations were quantified using an Enhanced BCA Protein Assay Kit (NCM, Suzhou, China). Equal amounts of protein (30 µg per lane) were resolved on 10% SDS-PAGE and transferred onto PVDF membranes (Immobilon-P, 0.22 μm; Merck Millipore). Membranes were blocked for 1 h at room temperature in TBS-T (Tris-buffered saline containing 0.1% Tween-20) supplemented with 5% non-fat dry milk. Primary antibody incubation was performed overnight at 4 °C using the following antibodies: rabbit anti-ATP5F1A (1:1000; Abcam, ab176569), mouse anti-DYKDDDDK Tag (1:1000; Proteintech, 66008-4), rabbit anti-LC3B (1:2000; Abcam, ab192890), rabbit anti-P62 (1:1000; Abcam, ab207305), and mouse anti-GAPDH (1:5000; Servicebio, GB15002-100). Following primary antibody incubation, membranes were washed three times with TBS-T and then incubated for 1 h at room temperature with HRP-conjugated secondary antibodies: goat anti-rabbit IgG (1:15000; Abcam, ab97051) and goat anti-mouse IgG (1:5000; Abclonal, AS003). After extensive washing with TBS-T, protein bands were visualized using Ultra-Sensitive ECL Chemiluminescent Substrate Kit (Biosharp, Beijing, China) and detected with the Tanon imaging system. Densitometric analysis of protein bands was performed using ImageJ software (National Institutes of Health, USA). Immunofluorescence HEK 293T cells were grown on coverslips and transfected as described. Cells were washed with PBS and fixed with 4% paraformaldehyde in PBS for 15 min at room temperature. Following fixation, cells were washed three times with PBS and permeabilized with 0.5% Triton X-100 in PBS for 15 min. After blocking with 1% bovine serum albumin for 1 h at room temperature, cells were incubated with primary antibodies overnight at 4 °C: anti-IMMT (1:500; Proteintech, 68226-1) and anti-DYKDDDDK tag (1:500; Proteintech, 80801-2). Subsequently, cells were washed three times and incubated with fluorophore-conjugated secondary antibodies for 1 h at room temperature in the dark: goat anti-mouse Multi-rAb CoraLite^® Plus 555 (1:500; Proteintech, RGAM003) and goat anti-rabbit Alexa Fluor^® 647 (1:500; Abcam, ab150079). Nuclei were counterstained with DAPI, and coverslips were mounted using anti-fade mounting medium (Beyotime, Shanghai, China). Images were acquired using a confocal microscope (Nikon AXR, Japan). Zebrafish maintenance and microinjection Wild-type (AB) and Tg (mnx1: eGFP) transgenic lines of zebrafish were raised at the Zebrafish Technology Platform of Southwest Medical University. All zebrafish were maintained at 28 °C with a 14 h light and 10 h dark cycle. The atp5fa1-e3i4-MO sequence (5′-GCCTAGAAGTTTGTCCCTCACCTTC-3′) and the standard control MO sequence (5′-CCTCTTACCTCAGTTACAATTTATA-3′) were synthesized by GeneTools (Philomat, OR, USA). According to standard procedures, 2 ng/nL was injected into the yolk pole of 300 zebrafish embryos at the one-cell stage. Microinjected embryos were placed in E3 medium (5 mM NaCl, 0.17 mM KCl, 0.33 mM CaCl[2], 0.33 mM MgSO[4] and 0.00001% methylene blue) and cultured in a 28.5 °C incubator. Zebrafish embryos used for fluorescence imaging were treated with E3 medium containing 0.05 mmol/L 1-phenyl-2-thiourea (PTU) at 24hpf to inhibit skin melanin production. Behavioral recording The zebrafish larvae were tested for their motor behavior using a zebrafish behavior analysis system (DanioVision, Noldus, Netherlands). A clean 24-well plate was used with 500 uL of culture medium per well and one 120hpf larva was tested (three independent experiments, with 8 larvae per group). Each test of dark field free-swimming was preceded by 10 min of adaptation and 30 min of tracking. The alternating light and dark test consisted of: (1) a 10-minute acclimatization period, (2) a sudden light change test, alternating between 10 min of bright light (white LED, ~ 1900 lx) and 10 min of dark (0 lx), totaling 60 min. Data were recorded as travel distance (cm) and speed (cm/min). Data were analyzed using EthoVision XT software. Transmission electron microscopy (TEM) Zebrafish larvae were pre-fixed with 3% glutaraldehyde and then post-fixed with 1% osmium tetroxide. Samples were then dehydrated through a graded acetone series (30%, 50%, 70%, 80%, 90%, 95%, and 100%, with three changes at 100%). Following dehydration, specimens were infiltrated with acetone and Epon-812 resin mixtures at ratios of 3:1, 1:1, and 1:3, then embedded in pure Epon-812 resin. Ultrathin Sects. (60–90 nm) were prepared using an ultramicrotome and collected on copper grids. Sections were double-stained with uranyl acetate (10–15 min) followed by lead citrate (1–2 min) at room temperature. The ultrastructural observation and analysis of mitochondria in zebrafish larvae neurons were conducted using a JEM-1400FLASH transmission electron microscope (JEOL, Tokyo, Japan). Mitochondrial complex V enzyme activity assay The mitochondrial complex V enzyme activity was determined using the mitochondrial respiratory chain complex V/ATP synthase activity detection kit (Solarbio, Beijing, China). Zebrafish larvae at 72 hpf (n = 15 larvae per group) were homogenized in cold isolation buffer, and mitochondria were extracted according to the manufacturer’s instructions. The assay principle involves complex V hydrolyzing ATP to produce ADP and Pi, with the rate of Pi increase measured colorimetrically at 340 nm. The calculation of complex V activity was normalized by protein concentration. RNA sequencing analysis Transcriptome RNA sequencing (RNA-seq) experiments were performed on 72hpf larvae in the control and experimental groups. There were 4 samples in each group, and 15 larvae in each sample. After washing, the larvae were snap-frozen in liquid nitrogen and sent to a commercial supplier for RNA extraction and high-throughput sequencing. The sequencing results were analyzed and plotted on the Majorbio cloud platform ([53]https://cloud.majorbio.com/). |log2FC| ≥ 1 and adjusted P < 0.05 were the screening criteria. In GO enrichment analysis and KEGG PATHWAY enrichment analysis, enrichment was considered significant when the adjusted P value (FDR) was < 0.05. mRNA microinjection For the rescue experiment, wild-type human ATP5F1A mRNA was synthesized using the mMESSAGE mMACHINE T7 Ultra Kit (Thermo Fisher, AM1345) according to the manufacturer’s instructions. Linearized plasmid DNA (plasmid construction described in Sect. [54]2.4) was used as a template for in vitro transcription, followed by DNase treatment and lithium chloride purification. The quantitative analysis was performed using NanoDrop. The mRNA was diluted to 200 ng/µL in 0.5× Danieau solution containing 0.05% phenol red as a tracer. Morpholino oligonucleotides and mRNA were co-injected into one-cell stage embryos. After injection, the embryos were transferred to E3 medium and incubated at 28.5 °C. Statistical analysis Analyses were performed using GraphPad Prism 9 software. Data normality was assessed using Shapiro-Wilk test. Statistical significance was calculated using Student t-test or analysis of variance (ANOVA), as appropriate. Data are presented as mean ± standard deviation (SD). Asterisks indicate the level of statistical significance, * indicates P < 0.05, ** indicates P < 0.01, *** indicates P < 0.001 and **** indicates P < 0.0001. Results Clinical presentation and laboratory findings The proband was a 2 years and 4 months oldfemale, the first child of non-consanguineous parents (Fig. [55]1A). She was referred to our pediatric rehabilitation clinic at 1 year and 10 months old due to global developmental delay. Physical examination revealed growth retardation, generalized hypotonia, markedly weakened support strength, inability to maintain lower limb extension, and impaired standing balance. Biochemical analysis demonstrated elevated serum lactate (3.05 mmol/L; normal: 0.5–2.2 mmol/L) and lactate dehydrogenase (LDH: 268.2 U/L; normal: 120–250 U/L). Routine blood tests, hepatic/renal function panels, and creatine kinase levels were within normal ranges. Metabolic screening via liquid chromatography-tandem mass spectrometry (LC-MS/MS) showed no significant abnormalities in amino acid or acylcarnitine profiles. Brain and full-spine MRI revealed no structural anomalies. Video electroencephalography (VEEG) during wake-sleep cycles detected intermittent 2.8–3.5 Hz slow-wave bursts localized to bilateral fronto-central-parietal regions, with right hemispheric predominance. At 2-years-and-4-month-old, motor activity was limited to crawling and supported standing, with independent ambulation unachieved. Speech development was restricted to monosyllabic utterances. Gesell Developmental Schedules evaluation demonstrated profound delays across domains: The developmental quotient (DQ) of adaptation, gross motor and fine motor was 29.0 (equivalent to 8.4 months); the DQ of language was 28.0 (equivalent to 8.1 months); and the DQ of personal-social behavior was 27.0 (equivalent to 7.5 months). Sensory evaluation revealed severe proprioceptive and balance deficits. Respiratory chain enzyme analysis was deferred due to parental refusal of muscle biopsy. Fig. 1. [56]Fig. 1 [57]Open in a new tab Identification of a de novo ATP5F1A mutation. A Pedigree chart. Arrow indicates the proband (II-1). The solid symbol indicates the affected individual (square for males and round for females). B Sanger sequencing confirming the de novo heterozygous c.1252G > A (p. Gly418Arg) variant in ATP5F1A ([58]NM_001001937.2). (C) Evolutionary conservation analysis of glycine-418 demonstrates complete conservation at this residue position across vertebrate species. (D) Structural simulation of ATP5F1A wild-type and mutant(p. Gly418Arg)protein. Models generated using SwissModel and visualized with PyMOL Whole exome sequencing identified a heterozygous c.1252G > A (p. Gly418Arg) variant in ATP5F1A ([59]NM_001001937.2), located in exon 10. Sanger sequencing confirmed this as a de novo mutation, absent in both parents (Fig. [60]1B). The variant was unreported in gnomAD population databases and submitted to ClinVar (accession: VCV002572074.1; [61]https://www.ncbi.nlm.nih.gov/clinvar). Evolutionary conservation analysis demonstrated high conservation of glycine at ATP5F1A position 418 in several species (Fig. [62]1C). Structural modeling revealed significant conformational alterations in the mutant protein: substitution of non-polar glycine with positively charged arginine at residue 418 introduced novel hydrogen bonding with valine-417 (Fig. [63]1D), potentially disrupting ATP synthase α-subunit folding. Bioinformatics prediction software unanimously supported pathogenicity: SIFT score = 0, damaging; PolyPhen-2 score = 1, probably damaging; MutationTaster score = 0.999, disease-causing. Following ACMG guidelines, this variant was classified as pathogenic. ATP5F1A mutations and human disease phenotypes This study summarizes all reported ATP5F1A variants that lead to human diseases (Table [64]1). A total of seven mutation sites were identified, including six missense mutations and one frameshift mutation (Fig. [65]2A). Patients inherit these mutations in both autosomal recessive (homozygous) and autosomal dominant (de novo heterozygous) patterns. Variants are distributed in both the nucleotide-binding domain and the C-terminal catalytic domain of ATP5F1A (Fig. [66]2B). Conservation analyses across species showed that all missense mutations occurred at highly conserved loci with a ConSurf score of at least 7 (Fig. [67]2C). Table 1. Clinical phenotype and genetic characteristics of ATP5F1A related diseases patients Patient Sex Age at Onset Clinical feature Variants Zygosity ETC biochemistry prognosis CI CII CIII CIV CV P1 M 18d failure-to-thrive, chronic diarrhoea, anaemia, hyperammonemia, lactic acidemia c.620 G>A(p.Arg207His) heterozygous NR normal normal normal ↓ alive(symptom resolution at12m) P2 F 9d failure-to-thrive, hyperammonemia, lactic acidemia c.620 G>A(p.Arg207His) heterozygous NR NR NR NR ↓ alive(symptom resolution at10m) P3 F 13d failure-to-thrive, chronic diarrhoea, anaemia, hyperammonemia, lactic acidemia, encephalopathy c.620 G>A(p.Arg207His) heterozygous normal normal normal normal ↓ alive(symptom resolution at15-18m) P4 F Newborn failure to thrive, microcephaly, encephalopathy, IUGR, hypotonia, pulmonaryhypertension, heart failure c.926 A>G(p.Tyr321cys) homozygous normal normal normal ↓ NR died at 3m P5 F Newborn NR c.926 A>G(p.Tyr321cys) homozygous normal normal ↓ ↓ NR died at 15m P6 NR Newborn intractable seizures, encephalopathy, high-pitched cry, horizontal and vertical nystagmus, abnormal primitive reflexes, tonus dysregulation c.985 C>T(p.Arg329cys) heterozygous normal normal normal normal ↓ died in the first weeks of life P7 NR Newborn c.985 C>T(p.Arg329cys) heterozygous normal normal normal normal ↓ died in the first weeks of life P8 F Early infancy developmental delay, failure-to-thrive, lactic acidosis c.620 G>A(p.Arg207His) heterozygous normal normal normal normal ↓ alive(symptom resolution) P9 M NR psychomotor delay, intellectual disability, ataxia, spastic paraparesis, dystonia c.545 G>A(p.Arg182Gln) heterozygous NR NR NR NR NR alive P10 F NR psychomotor retardation, spastic tetraparesis, generalized dystonia, absent speech, swallowing problems, increased blood lactate concentrations c.1037 C>T(p.Ser346Phe) heterozygous NR NR NR NR NR alive P11 M NR developmental delay, dystonia, cognitive impairment, myoclonus, pyramidal syndrome c.1404del (p.Glu469Serfs*3) heterozygous NR NR NR NR ↓ alive P12 F NR spastic gait impairment, mild intellectual disability c.1252G>A (p.Gly418Arg) heterozygous NR NR NR NR NR alive P13 F NR spastic paraparesis, mild neurodevelopmental impairments, delayed language development, borderline intellectual disability, autism spectrum disorder c.1252G>A (p.Gly418Arg) heterozygous NR NR NR NR NR alive P14 F NR bilateral lower-extremity spasticity c.1252G>A (p.Gly418Arg) heterozygous NR NR NR NR NR alive [68]Open in a new tab Abbreviations: ETC, electron transport chain; CI, complex I; CII, complex II; CIII, complex III; CIV, complex IV; CV, complex V; NR, not report; IUGR, intrauterine growth restriction Fig. 2. [69]Fig. 2 [70]Open in a new tab Distribution and conservation analysis of ATP5F1A pathogenic variants. A) Spatial distribution of 7 variants in the three-dimensional structure of human ATP5F1A protein. B Distribution of mutation sites in the ATP5F1A protein domains. Domain definitions are based on the InterPro database: ATP-synt_ab_N (β-barrel domain, InterPro: IPR000568), F1-ATPase_alpha_CD (nucleotide binding domain, InterPro: IPR000793), ATP-synt_ab_C (C-terminal catalytic domain, InterPro: IPR000194). Generated using IBS 2.0. C Conservation analysis of multiple sequences of ATP5F1A protein. The black line indicates the ConSurf conservation score (based on UniRef90 alignment, Bayesian algorithm), with a score of 1–9 (gradient blue-red, 1 = variable, 9 = highly conserved). Mutation sites are marked with vertical lines ATP5F1A deficiency encompasses a spectrum of disease severities. Based on clinical severity and prognosis, the phenotypes can be categorized into three groups: self-resolving transient neonatal metabolic dysfunction, persistent neurodevelopmental disorders accompanied by motor abnormalities, and severe early-onset mitochondrial encephalopathy that can lead to fatal consequences. Movement disorders, particularly dystonia, are the most common phenotype among these patients, followed by neurodevelopmental abnormalities and metabolic disturbances, including lactic acidosis and hyperammonemia. Functional studies on ATP5F1A mutants demonstrated varying degrees of ATPase dysfunction. The p.Arg207His mutant, associated with a transient neonatal phenotype, showed reduced enzyme activity [[71]14], while the p.Tyr321Cys mutant, associated with lethal encephalopathy, showed severe ATPase deficiency and mitochondrial DNA depletion [[72]15]. Recent investigations confirmed that the p.Glu469Serfs*3 frameshift mutation resulted in significantly reduced ATP5F1A mRNA levels and compromised ATPase complex assembly [[73]13]. This suggests that loss of ATP5F1A function could be a fundamental mechanism underlying ATP5F1A-related mitochondrial disorders. The ATP5F1A c.1252G > A mutation results in reduced mRNA and protein expression without altering mitochondrial localization To investigate the effects of the monoallelic missense mutation of ATP5F1A, wild-type (WT) and c.1252G > A mutant-type (MT) plasmid were constructed with FLAG tags. qPCR and Western blotting analysis demonstrated moderately but significantly reduced mRNA and protein expression levels in mutant-transfected cells compared to wild-type controls (Fig. [74]3A-C), suggesting that the point mutation leads to a decrease in protein expression. To clarify whether the ATP5F1A mutant protein changes its subcellular localization, immunofluorescence analysis showed that exogenously expressed ATP5F1A co-localized with the mitochondrial inner membrane protein IMMT in both wild-type and mutant transfected HEK 293T cells (Fig. [75]3D). ATP5F1A is localized to the mitochondrial inner membrane, while the p. Gly418Arg mutation does not change its mitochondrial localization. Fig. 3. [76]Fig. 3 [77]Open in a new tab Effect of p. Gly418Arg mutation on ATP5F1A mRNA and protein expression and subcellular localization. A. Relative expression level of ATP5F1A mRNA after HEK 293T cells were transfected with empty vector (NC), wild-type (WT) or mutant-type (MT, p. Gly418Arg) vector (qPCR detection, internal reference gene: GAPDH; data mean ± SD, n=3; **P<0.01, ****P<0.0001, one-way ANOVA). B. Immunoblotting analysis of ATP5F1A-FLAG protein (anti-FLAG antibody, internal reference: GAPDH). Representative images show the protein expression levels after empty vector, WT and MT transfection. C. Quantitative analysis of ATP5F1A-FLAG protein expression (ImageJ normalized to GAPDH; mean ± SD, n=3; **P<0.01, one-way ANOVA). D. Subcellular localization of ATP5F1A-FLAG in HEK 293T cells. Wild-type (WT) and mutant-type (MT) proteins (green, anti-FLAG antibody), mitochondrial inner membrane marker IMMT (red, anti-IMMT antibody) and cell nuclei (blue, DAPI staining). Scale bar, 10μm. ATP5F1A deficiency causes growth retardation in zebrafish To investigate the in vivo functional significance of ATP5F1A, we employed zebrafish (Danio rerio) as a model system. The zebrafish genome contains an orthologous atp5fa1 sharing 79.2% protein sequence homology with human ATP5F1A. Developmental expression profiling by RT-qPCR revealed atp5fa1 was highly expressed maternally in the zygote, decreased from 0 to 12hpf, and then gradually increased during the organogenesis period (12-120hpf) (Fig. [78]4A). The atp5fa1 mRNA and protein levels were significantly reduced in the zebrafish model after MO injections compared to control-MO injected larvae, confirming partial knockdown of atp5fa1. While the protein reduction was modest (30%), significant developmental phenotypes were observed (Fig. [79]4B, C and D), suggesting that even partial Atp5fa1 reduction can impact embryonic development. atp5fa1 knockdown in early embryos causes a sharp drop in embryo survival within 24hpf, and almost all embryos die at 144hpf (Fig. [80]4E). Morphometric analysis at 72hpf and 120hpf revealed a significant reduction in body length, accompanied by abnormal phenotypes such as body axis bending and pericardial edema (Fig. [81]4F and G). Fig. 4. [82]Fig. 4 [83]Open in a new tab atp5fa1 knockdown causes growth and development disorders in zebrafish. A Temporal expression profile of atp5fa1 mRNA during zebrafish embryonic development (qPCR detection, internal reference gene: β-actin). B The atp5fa1 mRNA expression levels in the control (control-MO) and atp5fa1-KD (atp5fa1-MO) larvae at 24hpf, 72hpf and 120hpf (mean ± SD, n = 15 embryos/group; ****P < 0.0001, Student t-test). C Immunoblotting analysis of Atp5fa1 protein (anti-ATP5F1A antibody, internal reference: Gapdh). Representative results show the difference in protein expression between the control group and the knockdown group. D Quantitative analysis of Atp5fa1 protein expression (ImageJ software normalized to Gapdh; mean ± SD, n = 3 independent experiments; ***P < 0.001, Student t-test). E Survival curves of control group and atp5fa1-KD group larvae (0-144hpf; total sample size n = 300 embryos/group). F Phenotypic comparison of 72hpf and 120hpf larvae. Scale bar, 300 μm. G Quantification of body length of 72hpf and 120hpf larvae (mean ± SD, n = 10; ****P < 0.0001, Student t-test) ATP5F1A deficiency causes abnormal neuronal development and motor dysfunction in zebrafish To explore the impact of ATP5F1A deficiency on motor function, the mnx1:eGFP transgenic strain of zebrafish was selected for research on motor neurons and axons. Comparative analysis revealed that the axon length of motor neurons in atp5fa1-knockdown zebrafish was shortened, with some axons completely absent (Fig. [84]5A). In rescue experiments using human wild-type ATP5F1A mRNA, although the distribution of motor neuron axons remained disordered, no shortening or absence of axons was observed (Fig. [85]5A). These results indicated that reduced atp5fa1 expression will lead to impaired development of motor nerves. Subsequently, behavioral tests on 120 hpf larvae were conducted (Fig. [86]5B). The free-swimming experiment showed that the total distance traveled and swimming speed of the atp5fa1-MO group were significantly lower than those of the control group, while the rescue experimental group partially restored motor function (Fig. [87]5C and D). In the 10-min light-dark alternation experiment of zebrafish, the juvenile fish in the atp5fa1-MO group showed no obvious response to the light stimulation, whereas mRNA rescue treatment partially recovered the light-dark responsive swimming behavior (Fig. [88]5E). Fig. 5. [89]Fig. 5 [90]Open in a new tab Effects of Atp5fa1 reduction on zebrafish motor function. A Representative images of peripheral neurons in transgenic (mnx1:mGFP) zebrafish at 72hpf. Scale bar, 200 μm. B Movement trajectories of 120hpf zebrafish larvae in the control group (control-MO), atp5fa1 knockdown group (atp5fa1-MO), atp5fa1-MO and ATP5F1A mRNA co-injection group (n = 8 embryos/group). The lines indicate movement trajectories. C Quantification analysis of total swimming distance. (Mean ± SD, n = 8 embryos/group; ****P < 0.0001, Student t-test). D Quantification analysis of velocity. (Mean ± SD, n = 8 embryos/group; ****P < 0.0001, Student t-test). E Curves showing changes in swimming distance over time for zebrafish larvae in the control group, atp5fa1-MO group and atp5fa1-MO group + ATP5F1A mRNA during the light-dark alternation experiment. Swimming distance represents cumulative distance recorded every 5 min. ATP5F1A deficiency causes impaired mitochondrial structure and function ATP5F1A is localized in the inner mitochondrial membrane, and it is worth investigating whether its deficiency affects the morphological structure and function of mitochondria. Therefore, after knocking down atp5fa1 using MO, the ultrastructure of mitochondria in the neurons of zebrafish larvae was observed through transmission electron microscopy. Compared to the control-MO group, the mitochondria in the atp5fa1-MO group showed a shallower matrix, and the mitochondrial cristae were shorter or even absent (Fig. [91]6A). At the same time, we explored the effect of Atp5fa1 reduction on the activity of mitochondrial complex V. The results showed that the activity of complex V in the atp5fa1-MO group decreased significantly. Compared to the control-MO group, the activity of complex V decreased by approximately 30% (Fig. [92]6B). Fig. 6. [93]Fig. 6 [94]Open in a new tab Effects of Atp5fa1 reduction on mitochondrial structure and function in zebrafish larvae. A Ultrastructure of mitochondria in the neurons of zebrafish larvae of the control group (control-MO) and atp5fa1 gene knockdown group (atp5fa1-MO) under transmission electron microscopy (The red framed area is a local magnified view, and the arrow indicates the disordered cristae structure or vacuolization; scale bar: 2 μm in the main image, 500 nm in the magnified image). B Comparison of the activity of mitochondrial complex V in zebrafish between the control group and atp5fa1-MO group (Mean ± SD; ***P < 0.001, Student t-test) Differential expression and enrichment analysis of mRNAs To determine the molecular mechanisms underlying the phenotypes of developmental disorders and motor dysfunction in zebrafish caused by ATP5F1A deficiency, this study performed RNA-seq on the whole embryos of control-MO and atp5fa1-MO zebrafish, and conducted a comparative analysis of the obtained data. Pearson correlation analysis was carried out based on the FPKM (Fragments Per Kilobase of exon model per Million mapped fragments) distribution in all samples. The results showed that the correlation coefficients of the samples within each group were all close to 1, indicating a strong correlation among the biological replicate samples, which ensured the reliability of the experimental data (Fig. [95]7A). Compared with the control group, a total of 2261 DEGs were identified in the atp5fa1 knockdown group. Among them, 562 genes were upregulated and 1699 genes were downregulated. The differentially expressed genes were visualized using a volcano plot and a bar chart (Fig. [96]7B). GO enrichment analysis showed that the differentially expressed genes generated by atp5fa1 knockdown were enriched in functions related to neurotransmission, including nervous system process, anterograde trans-synaptic signaling, chemical synaptic transmission, trans-synaptic signaling, regulation of trans-synaptic signaling, postsynaptic membrane, synaptic membrane, neurotransmitter receptor activity, etc. (Fig. [97]7C). qPCR verification showed that the mRNA levels of neural-related genes such as gabra1, grin1a, gad1b, syngr1a, gria3a, grm1a, hcn1 and syt16 were significantly decreased (Fig. [98]7E), suggesting that atp5fa1 plays an important role in maintaining neurotransmission function. Fig. 7. [99]Fig. 7 [100]Open in a new tab RNA-seq analysis of zebrafish model reveals the molecular regulatory network of atp5fa1 knockdown. A Transcriptome correlation analysis between samples (Pearson correlation coefficient heat map based on FPKM value). B Volcano plot of differentially expressed genes between control-MO group and atp5fa1-MO group. C Gene Ontology (GO) enrichment analysis of differentially expressed genes. D Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis of differentially expressed genes between the control-MO group and the atp5fa1-MO group. E qPCR validation of neurotransmission-related genes (gabra1, grin1a, gad1b, gria3a, grm1a, hcn1, syngr1a, syt16) (mean ± SD, n = 15 embryos/group; **P < 0.01, *** P < 0.001, ****P < 0.0001, Student t-test). F qPCR validation of Apelin-autophagy relative genes apln, becn1, and map1lc3b (mean ± SD, n = 15 embryos/group; **P < 0.01, ***P < 0.001, ****P < 0.0001, Student t-test). G) Western blot analysis of autophagy-related proteins P62, Lc3b-I, and Lc3b-II in control-MO and atp5fa1-MO zebrafish larvae at 72hpf. H Quantitative analysis of Lc3b-II protein expression normalized to Gapdh (mean ± SD, n = 3; **P < 0.01, Student t-test). I Quantitative analysis of P62 protein expression normalized to Gapdh (mean ± SED, n = 3; ***P < 0.001, Student t-test) After microinjecting Morpholino, 31 KEGG pathways were significantly enriched, including Neuroactive ligand-receptor interaction, PPAR signaling pathway, Apelin signaling pathway, and Calcium signaling pathway, etc. (Fig. [101]7D). Transcriptome sequencing showed that compared with the control-MO group, the differentially expressed genes in the atp5fa1-MO group were mainly enriched in the Apelin signaling pathway. To investigate the effect of Atp5fa1 reduction on the Apelin signaling pathway, qPCR analysis revealed that the mRNA expression level of apln was decreased in the atp5fa1-MO zebrafish model (Fig. [102]7F). Meanwhile, we found that the decrease in apln induced a reduction in the mRNA expression levels of becn1 and map1lc3b in the atp5fa1-MO zebrafish model (Fig. [103]7F). Western blot analysis was performed to detect the expression levels of autophagy-related proteins in zebrafish models (Fig. [104]7G). The results showed that the expression level of autophagy substrate protein P62 was significantly upregulated in the atp5fa1-MO group (Fig. [105]7H), while the expression of lipidated LC3-II was significantly downregulated (Fig. [106]7I). These changes indicate that the knockdown of atp5fa1 impairs the initiation of autophagy, while also reducing the efficiency of autophagic clearance. Discussion This study provides further evidence that the de novo ATP5F1A c.1252G > A (p.Gly418Arg) variant causes mitochondrial complex V deficiency, enhancing our understanding of its pathogenic mechanisms. The disease phenotype and underlying mechanisms of ATP5F1A deficiency were investigated using in vitro cell experiments and Atp5f1a-deficient zebrafish models. Mutations in the ATP5F1A gene result in disease phenotypes with significant clinical heterogeneity. Developmental delay, hyperlactatemia, and motor dysfunction were the most common features of ATP5F1A deficiency. Recently, Harrer et al. reported three patients from two families with hereditary spastic paraplegia (HSP) carrying the ATP5F1A c.1252G > A (p.Gly418Arg) variant, and for the first time, described the clinical features using protein structural modeling [[107]13]. This study independently identified the same variant in a Han Chinese patient, further confirming its pathogenicity and expanding the ethnic spectrum of affected individuals. Notably, while Harrer et al. primarily observed HSP, previously reported patients with ATP5F1A mutations—including this case—exhibited multisystem involvement, such as growth retardation, movement disorders, and metabolic abnormalities. This heterogeneity may arise from three factors: (i) genetic modifiers, which significantly influence disease severity through background genomic variation; (ii) ethnic-specific differences, reflecting the impact of genetic background on phenotypic expression; (iii) epigenetic regulation, such as DNA methylation, which may modulate phenotype expression [[108]16]. Although the exact mechanisms of ATP5F1A modification remain unclear, similar genetic screening models have demonstrated that modifier factors can shape mutant phenotypes, offering important insights into phenotypic variability. Zebrafish is a valuable model organism for studying human genetic diseases due to its optical transparency in embryos and larvae, ease of genetic manipulation, and high genetic homology with humans [[109]17]. In recent years, zebrafish models have successfully replicated key phenotypes of various mitochondrial complex deficiencies. Knockout of the ptpmt1 gene causes growth retardation, craniofacial abnormalities, and swim bladder loss, mimicking complex IV deficiency–related diseases [[110]18]. The chchd10 gene knockout results in motor coordination loss and disrupted neuromuscular synapses, reflecting primary mitochondrial disease features [[111]19]. The ndufb7-MO model exhibits ventricular dilation and lactic acidosis, indicating metabolic compensation for complex I dysfunction [[112]20]. In this study, the atp5fa1-MO zebrafish model showed growth retardation and impaired motor function, mirroring phenotypes observed in patients with ATP5F1A mutations. This model provides an effective tool to explore the molecular mechanisms and potential treatments for complex V deficiency. To rule out off-target effects of Morpholino technology, we performed rescue experiments by injecting human wild-type ATP5F1A mRNA. Partial recovery of abnormal motor neuron morphology confirmed a causal link between the gene defect and observed phenotypes. Mitochondrial function is essential for neural development [[113]21]. During neurogenesis, metabolism shifts from glycolysis to OXPHOS. In mature neurons, energy production relies mainly on OXPHOS to support complex neural activity [[114]22, [115]23]. Previous studies using mitochondrial gene-deficient models have shown that impaired OXPHOS leads to neurodevelopmental disorders (NDDs) [[116]24] and neurodegenerative diseases [[117]25]. NDUFB7 deficiency causes complex I dysfunction and reduced neuronal volume in the zebrafish midbrain and hindbrain [[118]20]. In FARS2-deficient mice, disrupted mitochondrial translation impairs OXPHOS, blocking ectoderm development and neurogenesis [[119]26]. In this study, atp5fa1 knockdown in zebrafish was used to investigate nervous system pathology under mitochondrial complex V deficiency. Results showed disorganized mitochondrial cristae and reduced complex V activity in neurons, along with abnormal neural development and motor impairment. These phenotypes closely match neurological symptoms in ATP5F1A-deficient patients. Transcriptome analysis revealed that atp5fa1 downregulation altered the expression of 29 mitochondrial-related genes, including those involved in ATP synthase, the electron transport chain, and carrier proteins. These findings suggest that mitochondrial dysfunction underlies the observed neuronal abnormalities. Therefore, reduced ATP5F1A expression may cause neurodevelopmental defects by disrupting mitochondrial structure and function. Transcriptomic analysis revealed a significant downregulation of the Apelin signaling pathway in atp5fa1 knockdown zebrafish models. Apelin (APLN), an endogenous ligand of the G protein-coupled receptor APJ, has a complex and context-dependent relationship with autophagy in neurodegenerative diseases. The precursor is enzymatically cleaved into several bioactive forms, including apelin-36, apelin-26, apelin-19, apelin-17, apelin-13, Pyr-apelin-13, and apelin-12 [[120]27, [121]28]. In Alzheimer’s disease models, Apelin-13 inhibits autophagy by activating the PI3K/Akt/mTOR pathway, reducing Beclin-1 and LC3B-II expression and exerting neuroprotective effects [[122]29, [123]30]. Conversely, in Parkinson’s disease models, Apelin activates autophagy via the AMPK/mTOR/ULK pathway to clear pathological protein aggregates [[124]31]. qPCR and Western blot analyses in this study showed that atp5fa1 knockdown significantly reduced apln and becn1 mRNA levels, decreased Lc3b-II protein expression, and increased P62 accumulation. Therefore, this coordinated pattern suggests that atp5fa1 knockdown may disrupt Apelin signaling and impair autophagy progression. Based on the observed mitochondrial dysfunction and autophagy dysregulation, we propose a comprehensive therapeutic strategy for ATP5F1A-related diseases. Modulating autophagy is a promising approach, and enhancers like rapamycin warrant systematic evaluation. Additionally, mitochondrial bioenergetics enhancers like coenzyme Q10, creatine, and nicotinamide riboside may help compensate for ATP deficiency [[125]32, [126]33, [127]34]. Drug development should prioritize patient-derived iPSC neuronal models for personalized screening [[128]35]. Furthermore, integrating CRISPR-Cas9 gene editing offers opportunities for mechanistic studies and potential curative therapies by correcting pathogenic mutations [[129]36]. This study has several limitations. Functional analysis was conducted in HEK 293T cells, which do not fully replicate affected tissue environments; patient-derived cells would provide more relevant models. While MO-mediated knockdown in zebrafish effectively simulates the heterozygous phenotype, it carries risks of off-target effects and transient action. Validation using stable CRISPR/Cas9 knockout lines would strengthen these findings. Single-cell multi-omics approaches are essential to unravel molecular complexity, as integrating transcriptomics, proteomics, and metabolomics can identify cell type-specific vulnerabilities and novel therapeutic targets within dysregulated networks [[130]37] thereby facilitating translation from mechanistic insight to precision therapy. In summary, this study preliminarily elucidates the impact of ATP5F1A deficiency on clinical phenotype, biological function, and molecular mechanisms through clinical data, genetic analysis, and experiments using cell and animal models. It confirms p.Gly418Arg as a recurrent pathogenic ATP5F1A mutation and establishes a link between reduced ATP5F1A expression and neurological phenotypes. Our findings suggest that the apelin–autophagy signaling axis may serve as a therapeutic target in ATP5F1A-related disorders, offering new mechanistic insights for targeted treatment development. Acknowledgements