Next Article in Journal
Brain Monoamine Deficits in the CD Mouse Model of Williams–Beuren Syndrome
Previous Article in Journal
Angiotensin-Converting Enzyme Inhibitors and Metabolic Aging: A Drosophila Perspective
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Transcription, Maturation and Degradation of Mitochondrial RNA: Implications for Innate Immune Response

1
National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China
2
Department of Pathology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
*
Authors to whom correspondence should be addressed.
Biomolecules 2025, 15(10), 1379; https://doi.org/10.3390/biom15101379
Submission received: 31 July 2025 / Revised: 19 September 2025 / Accepted: 24 September 2025 / Published: 28 September 2025
(This article belongs to the Special Issue Mitochondria as a Target for Tissue Repair and Regeneration)

Abstract

Mitochondria are crucial for a wide range of cellular processes. One of the most important is innate immunity regulation. Apart from functioning as a signaling hub in immune reactions, mitochondrial nucleic acids can themselves act as damage-associated molecular patterns (DAMPs) to participate in immune processes directly. This review synthesizes the current understanding of mitochondrial RNA (mtRNA) biology and its link to immune activation through aberrant accumulation. We focus on its origin through bidirectional mitochondrial transcription and metabolism, encompassing maturation (cleavage, polyadenylation, modification) and degradation. Dysregulation of mtRNA metabolism leads to mt-dsRNA (mitochondrial double-stranded RNA) accumulation, which escapes mitochondria via specific channels into the cytosol and serves as DAMPs to trigger an immune response. We discuss the critical roles of key regulatory factors, including PNPT1 (PNPase, Polyribonucleotide Nucleotidyltrans ferase 1), in controlling mt-dsRNA levels and preventing inappropriate immune activation. Finally, we review the implications of mt-dsRNA-driven inflammation in human diseases, including autoimmune disorders, cellular senescence, and viral infection pathologies, highlighting unresolved questions regarding mt-dsRNA release mechanisms.

1. Introduction

Mitochondria are ubiquitously distributed and indispensable intracellular organelles in eukaryotic cells. Mitochondria are semi-independent double membrane-bound organelles with their own DNA genome, known as mitochondrial DNA (mtDNA). The human mitochondrial genome is a 16,569 bp double-stranded circular DNA and is packaged by a ranged of proteins [1,2]. These two individual strands are referred to as heavy (H) strands and light (L) strands according to the differentiated guanine (G) content [3,4]. mtDNA encode a total of 37 genes, which include 22 transfer RNAs (tRNAs), 2 ribosomal RNAs (rRNAs), and 13 messenger RNAs (mRNAs) that specify core subunits of the OXPHOS (oxidative phosphorylation) system (Figure 1). Beyond their primary role as the major site of ATP generation through OXPHOS, mitochondria are also central to diverse cellular processes, including cell death pathways (apoptosis, pyroptosis, ferroptosis, necrosis, etc.), signal transduction, cell cycle regulation, cellular metabolism, and immune responses [5,6,7,8].
Mitochondria participate in immune responses in two distinct ways. On the one hand, they serve as a platform for antiviral signaling proteins in innate immune reactions. On the other hand, mitochondrial nucleic acids (mt-NAs) act as damage-associated molecular patterns (DAMPs) to engage in immune responses. Mitochondrial antiviral signaling proteins (MAVS), a key adapter protein in the innate immune response to RNA viruses, are located on the outer mitochondrial membrane. It serves as a signaling hub downstream of RIG-I-like receptors (RLRs). By activating interferon (IFN) and inflammatory cytokine pathways, MAVS coordinates the host’s antiviral defense [9,10,11,12,13]. Mitochondrial nucleic acids, including mitochondrial DNA (mtDNA) and mitochondrial RNA (mtRNA), are recognized as non-self-molecules. They function as immune activators, capable of triggering innate immune responses. When mitochondria are damaged by events like oxidative stress or viral infection, mtDNA can escape through several mechanisms, including BAX/BAK macropores, the mPTP (mitochondrial permeability transition pores) and VDAC (voltage-dependent anion channel), or mitochondrial-derived vesicles (MDVs). This released mtDNA then activates the immune response, including the cGAS–STING signaling pathway and the NLRP3 inflammasome [14,15,16,17,18]. Aberrantly accumulated mitochondrial RNA can also leak through pathways such as BAX/BAK and VDAC1, thereby activating the MDA5 (melanoma differentiation-associated gene 5)-driven antiviral signaling pathway and triggering a type I interferon response [19,20]. The mtDNA-cGAS-STING signal pathway is well documented. In this review, we focus on the mtRNA mediated immune responses and diseases caused by their abnormalities. To understand these processes, it is essential to first examine how mtRNA is processed—including transcription, splicing, modification, and maturation—as well as its degradation. These pathways operate in a coordinated manner, and disruptions at any stage can lead to mtRNA abnormalities such as accumulation, which may ultimately trigger immune activation. We will delineate the key steps in mtRNA biogenesis and turnover, and examine specific defects that contribute to mtRNA-driven inflammatory pathways.

2. Mitochondrial Transcription Process

Human somatic cells contain thousands of copies of mitochondrial DNA (mtDNA). Unlike nuclear gene expression, mtDNA expression is regulated through mechanisms controlling both the proportion of mtDNA molecules available for transcription and post-transcriptional processes. Mitochondrial transcription is driven by the mitochondrial RNA polymerase (POLRMT) [4,21]. However, POLRMT cannot initiate transcription without the essential factors TFAM (Transcription Factor A, Mitochondrial) and TFB2M (Transcription Factor B2, Mitochondrial). TFAM, the primary protein component of the mitochondrial nucleoid, crucial for mtDNA stability, plays a vital role in transcription initiation [22,23]. It binds to promoters on both the heavy-strand promoter (HSP) and light-strand promoter (LSP), induces DNA bending, and recruits POLRMT via its N-terminal domain to form a binary complex, thereby facilitating assembly of the transcription initiation complex [24,25]. Then, the TFB2M subsequently joins this complex. Functionally analogous to bacterial σ factors, TFB2M inserts into the active site of POLRMT to facilitate promoter melting [26,27]. The POLRMT-TFAM-TFB2M ternary complex catalyzes the formation of the first phosphodiester bond, generating short RNA transcripts (<10 nucleotides). During this initiation phase, POLRMT’s N-terminal domain maintains contact with the promoter, preventing premature complex dissociation [21]. Upon extension of the RNA to 15–20 nucleotides, steric pressure from the nascent RNA-DNA hybrid duplex displaces TFB2M from the polymerase active site [21]. Finally, the transcription elongation factor (TEFM) binds to the catalytic domain of POLRMT. By preventing premature separation of the RNA template strand, TEFM enhances the polymerase’s processivity by >100-fold, enabling POLRMT to transcribe the full 16 kb mitochondrial genome [27,28]. Mitochondrial transcription is terminated by MTERF1 (Mitochondrial Transcription Termination Factor 1) [29]. MTERF1 specifically recognizes the mitochondrial transcription termination sequence on mtDNA [30]. The crystal structure of human MTERF1 reveals that upon binding double-stranded DNA containing this sequence, the protein unwinds the DNA, flipping out three nucleotides critical for termination activity [30]. These flipped-out nucleotides interact with MTERF1 residues, stabilizing the flipped-out base conformation, which leads to dissociation of the POLRMT from the DNA template, thereby terminating transcription [31,32,33]. The transcription process of mitochondrial RNA is shown in the upper left corner of Figure 1.

3. Mitochondrial RNA Maturation

Mitochondrial RNA maturation is a complex and highly regulated process that converts polycistronic transcripts into functional mRNAs, tRNAs, and rRNAs. Unlike nuclear RNA processing, mitochondrial RNA maturation occurs in the mitochondrial matrix and involves multiple enzymatic steps, including endonucleolytic cleavage, post-transcriptional modifications (Figure 1) [34,35].

3.1. Cleavage of Polycistronic Transcripts

Mitochondrial RNA transcription proceeds bidirectionally from a single intergenic non-coding region (NCR), producing polycistronic RNAs that encompass almost the entire genome. These transcripts contain clusters of mRNAs, rRNAs, and tRNAs, which are flanked by tRNA sequences [36]. The “tRNA punctuation model” hypothesizes that tRNAs serve as recognition sites for endonucleases, guiding the cleavage of polycistronic RNAs into individual functional RNAs [37]. Endonucleases such as RNase P and RNase Z recognize specific tRNA sequences and structures, cleaving the precursor RNA at tRNA boundaries to release mature tRNAs, mRNAs, and rRNAs [38]. RNase Z (also known as ELAC2 in humans) is a metal-dependent endoribonuclease involved in the 3′-end processing of precursor tRNAs (pre-tRNAs) [39,40]. Its primary function is to remove trailer sequences from the 3′ end of pre-tRNAs, generating a terminus suitable for the subsequent addition of the conserved CCA trinucleotide [40,41]. RNase P is a ribonucleoprotein enzyme responsible for the 5′-end processing of pre-tRNAs. Its primary function is to catalyze the cleavage and removal of the 5′ leader sequence from pre-tRNAs, generating a tRNA molecule with a mature 5′ terminus. RNase P and RNase Z typically function sequentially in the tRNA maturation pathway. Their coordinated actions are essential for the complete processing and release of the mature tRNA molecule.
Mitochondrial RNase P is composed of three proteins: TRMT10C (TRNA Methyltransferase 10C, also called MRPP1), HSPD17B10 (Hydroxysteroid 17-Beta Dehydrogenase 10, also called MRRP2), and PRORP (Protein Only RNase P Catalytic Subunit, also called MRRP3) [38]. PRORP serves as the catalytic subunit of mitochondrial RNase P and belongs to the Protein-only RNase P (PRORP) family [42,43]. It is responsible for catalyzing the cleavage of the 5′ leader sequence of precursor tRNAs (pre-tRNAs), a critical step in tRNA maturation [41,44]. However, PRORP cannot exhibit RNase P activity by itself. Its efficient pre-tRNA cleavage function requires the accessory proteins TRMT10C and HSPD17B10. TRMT10C possesses methyltransferase activity and is involved in tRNA modification, specifically catalyzing the N1-methylation of purine bases within tRNA molecules [44,45]. HSPD17B10 forms a stable complex with TRMT10C [46]. Notably, the methyltransferase activity of TRMT10C is not required for supporting the cleavage activity of PRORP. Instead, the TRMT10C-HSD17B10 complex enhances cleavage efficiency primarily by promoting PRORP’s binding to its substrate [41,46]. While it is established that nuclear RNA modifications are co-transcriptionally incorporated, the temporal relationship between TRMT10C-mediated methylation and RNA cleavage during mitochondrial RNA processing remains unclear.
Some mitochondrial transcripts are not punctuated by tRNAs, such as between the genes ATP6/ATP8 and COX3. However, the mechanism by which their primary transcripts are processed into separate, mature mRNAs remains unclear. Recent studies have demonstrated that FAST kinase domain-containing protein 4 (FASTKD4) and FASTKD5 may be related to this process [47]. These findings highlight limitations of the tRNA punctuation model. Moreover, the recent identification of diverse types of mitochondrial non-coding RNAs (mt-ncRNAs), including long non-coding RNAs (lncRNAs), small non-coding RNAs (sncRNAs) and identified circular RNAs (circRNAs), further indicates that the tRNA punctuation model cannot fully account for all cleavage events involved in mitochondrial primary transcript processing or the biogenesis of all mitochondrial RNAs [48,49,50]. The mt-ncRNAs’ biogenesis relies on the mitochondrial transcription machinery, and subsequent processing. Their release from primary transcripts may occur through both punctuation-dependent and independent on “tRNA punctuation model”. For example, the maturation of lncRNAs, including lncCytb, lncND5, and lncND6, requires involvement of the mitochondrial RNase P protein 1 (MRPP1) for processing at both the 5′ and 3′ ends [48,51]. Mitochondrion-encoded circular RNAs (mecciRNAs) are formed through head-to-tail splicing of precursor RNAs [50]. Although their precise biogenic pathways are still under active investigation, they are generally transcribed from mtDNA and cyclized within the mitochondria. The 7S RNA is transcribed from the non-coding D-loop region of mitochondrial DNA [52]. Its precursor is an independently transcribed ~180 bp RNA molecule that matures without requiring additional cleavage; instead, it only undergoes 3′ polyadenylation to form the functional transcript. The mature 7S RNA can bind to POLRMT and promote its homodimerization, thereby inhibiting mitochondrial transcription [52].

3.2. Mitochondrial RNA Post-Transcriptional Modifications

3.2.1. mt-mRNA Post-Transcriptional Modification

Post-transcriptional modifications are essential for mitochondrial RNA maturation and biofunction. Mitochondrial RNA modifications, although less numerous than their cytoplasmic counterparts, encompass several types and play crucial roles in precise protein synthesis [53]. The most common is the adenylation of the 3′-end mitochondrial mRNA. Except for MT-ND6, most mitochondrial mRNAs undergo 3′ polyadenylation after cleavage from the primary transcript [54,55]. This process is catalyzed by mitochondrial poly(A) polymerase (mtPAP, also known as PAPD1), a homodimeric enzyme [56,57]. For nuclear-encoded genes, polyadenylation plays a critical role in promoting mRNA stability and protein translation [58]. In contrast, in mitochondrial mRNA, polyadenylation functions not only to enhance mRNA stability but also to promote tRNA maturation [59]. The absence of mtPAP results in the loss of polyadenylation across all assayed mitochondrial transcripts, while oligoadenylation is preserved [60]. Importantly, the impact of this loss on transcript stability is transcript-dependent [61]. The other protein related to polyadenylation is LRPPRC (Leucine-Rich PPR Motif-Containing Protein). LRPPRC stabilizes mitochondrial RNA by binding to it and acting as a molecular chaperone [62]. It forms a complex with SLIRP (SRA Stem-Loop Interacting RNA Binding Protein) to maintain the secondary structures of mitochondrial mRNAs, preventing their degradation and ensuring their stability [63]. Studies demonstrate that loss of SLIRP protein significantly impairs the stability of LRPPRC protein [64]. This destabilization subsequently exerts detrimental effects on mitochondrial mRNA levels [64]. In addition, LRPPRC participates in regulating RNA polyadenylation and translation. A recent study reveals that the LRPPRC-SLIRP protein complex binds to mtRNA to stabilize it and expose initiation sites, bridges and positions the mitochondrial ribosomes, regulates translation initiation/elongation via interacting with mitochondrial translation factors, and ensures translation accuracy and mtRNA homeostasis [65]. Notably, mutations in the LRPPRC gene are conclusively linked to the development of Leigh syndrome (LSFC) [66]. The underlying mechanism involves LRPPRC mutations reducing mitochondrial mRNA levels, which compromises the proper assembly and function of cytochrome c oxidase (COX), ultimately leading to the pathogenesis of Leigh syndrome [66].
In addition to polycistronic cleavage and mRNA polyadenylation, base modifications are also part of the mitochondrial RNA maturation process. To date, over 170 chemical modifications have been identified in RNA, affecting the function of diverse RNAs, including nuclear and mitochondrial RNAs [67]. The most prevalent among these modifications in mitochondrial mRNA are methylation and pseudouridylation [68]. Methylation of mitochondrial mRNA (mt-mRNA) predominantly involves N1-methyladenosine (m1A), catalyzed by the mitochondrial methyltransferase complex TRMT61B (TRNA Methyltransferase 61B) and TRMT6/61A (TRNA Methyltransferase 6) [69]. This modification disrupts A-U base pairing and reduces mitochondrial translation efficiency. Pseudouridine (Ψ) modifications are putatively mediated by enzymes such as RPUSD3 (RNA Pseudouridine Synthase D3) and TRUB2 (RNA Pseudouridine Synthase Domain Containing 2), although their specific biological functions in mt-mRNA remain poorly characterized [70]. The m6A modification, ubiquitous in nuclear and cytosolic mRNAs, has also been detected in mt-mRNAs [71]. However, the methyltransferase(s) responsible for depositing m6A within mitochondria remain unidentified.

3.2.2. mt-tRNA Post-Transcriptional Modification

Among these three RNA types, tRNA exhibits the most abundant base modifications. Following its release from polycistronic RNA by RNase P and RNase Z cleavage, the 3′ end of mt-tRNA is matured through the addition of the universally conserved CCA (cytosine–cytosine–adenine) sequence by tRNA nucleotidyltransferase TRNT1 [45,72]. The CCA tail is crucial for tRNA aminoacylation, ribosomal binding, and protein synthesis [73]. Deficiencies in TRNT1 impair tRNA integrity and disrupt protein homeostasis [73]. Mutations or loss of TRNT1 function causes a multisystemic and clinically heterogeneous disease termed SIFD (sideroblastic anemia with B-cell immunodeficiency, periodic fevers, and developmental delay; SIFD) [74]. Additionally, methylation and pseudouridylation are widespread modifications on mitochondrial tRNAs (mt-tRNAs). Specifically, the adenine or guanine at position 9 is commonly methylated to m1A9 or m1G9, catalyzed by the TRMT10C-HSPD17B10 complex [41]. These modifications stabilize tRNA structure and function, thereby influencing protein translation [65]. TRMT10C is an S-adenosylmethionine (SAM)-dependent methyltransferase that catalyzes the transfer of a methyl group to the N1 position of adenine or guanine at position 9 (A9/G9) in mitochondrial tRNA [75]. In contrast, HSPD17B10 likely acts as a chaperone, stabilizing TRMT10C. Mutations in HSPD17B10 result in loss of TRMT10C and impaired processing of mitochondrial heavy-strand polycistronic transcripts [76]. m5C (5-methylcytosine) modification has also been detected at position 48–50 of mt-tRNA, which is added by NSUN2 (NOP2/Sun RNA Methyltransferase 2) [77]. TRMT61B was identified to be the methyltransferase of m1A at position 58 of several mt-tRNAs, which enhances the stability of the structure [78]. Pseudouridine (Ψ), one of the most abundant tRNA modifications and often termed the ‘fifth nucleotide’, is catalyzed by enzymes including PUS1 (Pseudouridine Synthase 1), RPUSD3, and TRUB2 (TruB Pseudouridine Synthase Family Member 2) [70,79,80]. PUS1 modifies uridine residues at positions 27 and 28 (U27/U28) in tRNA, whereas the specific modification sites for RPUSD3 and TRUB2 remain incompletely characterized [70,79]. Pseudouridine (Ψ) modifications in mt-tRNA contribute to structural stability and are essential for mitochondrial function [81]. Modifications within the anticodon loop of tRNA, particularly those at positions 34 and 37, are crucial for the fidelity and efficiency of codon-anticodon recognition. Among the diverse modifications found at position 37, m5C is relatively common. This methylation is catalyzed by the enzyme NSUN3 (NOP2/Sun RNA Methyltransferase 3), and can be reversed by the demethylase ALKBH1 (AlkB Homolog 1) [82,83]. Position 37 exhibits a remarkable variety of modification types. Key examples include m1G (1-methylguanosine), added by TRMT5 (TRNA Methyltransferase 5) [84]; i6A (N6-(dimethylallyl) adenosine), synthesized by TRIT1 (TRNA Isopentenyltransferase 1) [85]; and t6A (N6-threonylcarbamoyladenosine), whose formation requires the co-catalytic activity of YRDC (YrdC N6-Threonylcarbamoyltransferase Domain Containing) and OSGEPL1 (O-Sialoglycoprotein Endopeptidase Like 1) [86]. Collectively, these modifications at position 37 play a vital role in stabilizing the interaction between the codon and anticodon during translation.

3.2.3. mt-rRNA Post-Transcriptional Modification

Human mitochondrial rRNA, composed of the 16S and 12S large subunits, features three key modification types: 2′-O-ribose methylation, base methylation, and pseudouridylation. In the 16S rRNA, three specific 2′-O-ribose methylation sites exist (Gm1145, Um1369, and Gm1370) catalyzed by MRM1 (Mitochondrial RRNA Methyltransferase 1), MRM2 (Mitochondrial RRNA Methyltransferase 2), and MRM3 (Mitochondrial RRNA Methyltransferase 3), respectively; these modifications are essential for assembling the mitochondrial large ribosomal subunit and enabling mitochondrial protein translation [87,88]. Base methylation, specifically m1A, also occurs at position 947 and is catalyzed by TRMT61B [89]. Pseudouridylation at position 1397, mediated by RPUSD4 (RNA Pseudouridine Synthase D4), contributes to large subunit stability and promotes mitochondrial protein translation [70,90].
The 12S rRNA harbors abundant m4C and m5C base methylations. METTL15 (Methyltransferase 15) catalyzes m4C modification at position 839, NSUN4 (NOP2/Sun RNA Methyltransferase 4) mediates m5C modification at position 84, and METTL17 (Methyltransferase Like 17) facilitates both m4C modification at position 840 and m5C modification at position 842 [91,92,93]. The clustered methylations at positions 839–842 facilitate the translation of mitochondrial transcripts [91,92,93]. Loss of METTL17 significantly impairs mitochondrial protein translation and inhibits the mitochondrial oxidative respiratory chain; notably, METTL17 deficiency has also been reported to suppress tumor proliferation [93,94,95]. Similarly, NSUN4 deficiency in mice inhibits mitochondrial translation, while the absence of METTL15 disrupts mitochondrial ribosome assembly [92,96]. 12S rRNA also undergoes m62A dimethylation at positions 936 and 937, which is deposited by TFB1M (Transcription Factor B1, Mitochondrial) [97]. The m62A dimethylation modification promotes the binding of Ribosome Factor A (RBFA) to rRNA and is essential for ribosome assembly [98].

4. Mitochondrial RNA Degradation

Mitochondrial mRNAs typically have a half-life of 1–2 h [99]. The primary complex responsible for their degradation is the hSUV3/PNPase complex, within specialized D-foci in the mitochondrial matrix [99]. PNPase (also named PNPT1) provides 3′ → 5′ exoribonuclease activity, while the ATP-dependent helicase hSUV3 (Suv3 Like RNA Helicase) unwinds structured RNA substrates [99]. This complex preferentially degrades aberrant antisense mtRNAs and contributes to mRNA turnover, generating short oligonucleotides [99,100]. These products are subsequently processed to monoribonucleotides by the 3′ → 5′ exonuclease REXO2 (RNA Exonuclease 2) ensuring nucleotide recycling and preventing accumulation of inhibitory fragments (Figure 1) [101]. Degradation is dynamically opposed by the LRPPRC/SLIRP stabilization complex: LRPPRC and SLIRP cooperatively bind mt-mRNAs/rRNAs, suppress degradosome activity, prevent destructive secondary structures, and facilitate polyadenylation by mtPAP [102]. Critically, mtRNA stability is further enhanced through mitoribosome association; ribosomal binding physically shields transcripts from degradation, with depletion experiments confirming that unbound mRNAs are vulnerable to decay [103].
The degradosome also governs mitochondrial nucleic acid homeostasis beyond canonical decay. By degrading antisense transcripts, it limits mitochondrial double-stranded RNA (mt-dsRNA) accumulation. PNPase’s sublocalization dictates distinct roles: in the matrix (with SUV3), it degrades dsRNA, while in the intermembrane space (SUV3-independent), it sequesters dsRNA, preventing cytosolic leakage and inflammatory responses [104]. The RNA-binding protein GRSF1 (G-Rich RNA Sequence Binding Factor 1) enhances degradation specificity by resolving stable G-quadruplex (G4) structures in antisense RNAs via its quasi-RRM domains, enabling their unwinding and subsequent degradosome targeting—an adaptation to vertebrate G4-rich mitochondrial genomes. Dysregulation of this finely tuned system underlies severe pathologies. PNPase mutations (e.g., Gln387Arg, Glu475Gly) disrupt trimerization, impairing RNA import and causing early-onset neurological disorders (encephalopathy, hearing loss) [105,106]. LRPPRC mutations cause Leigh Syndrome French Canadian type (LSFC), a disorder characterized by the features described above. Notably, LRPPRC overexpression in diverse cancers promotes tumorigenesis by inhibiting apoptosis, enhancing invasion, and suppressing mitophagy through aberrant Parkin interaction [107,108]. Thus, precise mtRNA degradation is indispensable for mitochondrial integrity and cellular health, with defects impacting development, neurology, and oncology.

5. Mitochondrial RNA-Mediated Immunity

5.1. MAVS Signal Pathway

Mitochondria serve as signaling hubs for immune responses, among which the RNA-mediated MAVS signaling pathway is the most prominent. The MAVS signaling pathway is a pivotal innate immune defense mechanism that orchestrates the host response against viral infections. This intricate cascade is initiated upon the recognition of viral RNA by specialized pattern recognition receptors (PRRs) localized in various cellular compartments, including the cytoplasm and endosomes [109]. Key PRRs involved in this process include the retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs), such as RIG-I and melanoma differentiation-associated gene 5 (MDA5), as well as toll-like receptors (TLRs) like TLR3, TLR7, and TLR8 [110,111]. RIG-I selectively binds to short double-stranded RNA (dsRNA) with 5′ triphosphate ends, a molecular signature often presents in viral genomes or replication intermediates [112,113]. In contrast, MDA5 recognizes long dsRNA molecules, which are typical byproducts of viral replication [114]. TLR3, localized in endosomes, detects dsRNA, while TLR7 and TLR8 (expressed in immune cells like plasmacytoid dendritic cells) sense single-stranded RNA (ssRNA) from viruses [115,116].
Upon binding to viral RNA, these PRRs undergo conformational changes and activate downstream signaling. For RIG-I and MDA5, this activation involves the recruitment of the adaptor protein-MAVS, which is anchored to the mitochondrial outer membrane [11]. The interaction between activated RLRs and MAVS is mediated by homophilic caspase recruitment domain (CARD) interactions: RIG-I and MDA5 possess N-terminal CARDs that bind to the CARD of MAVS, triggering MAVS oligomerization [117,118].
MAVS oligomerization is a critical step that converts the initial RNA recognition event into a robust signaling response. Oligomerized MAVS forms large signaling complexes on the mitochondrial membrane, often referred to as “MAVS aggregates,” which serve as a platform for recruiting downstream signaling molecules [117]. These aggregates propagate the signal through two major pathways: the NF-κB (Nuclear Factor Kappa B Subunit 1)pathway and the interferon regulatory factor (IRF) pathway [117].
In the NF-κB pathway, MAVS recruits tumor necrosis factor receptor-associated factors (TRAFs), such as TRAF2, TRAF3, and TRAF6 [119,120,121]. TRAF6 activates the TAK1 (transforming growth factor-β-activated kinase 1) complex, which in turn phosphorylates IκB kinase (IKK) subunits [122]. Phosphorylation of IκB (inhibitor of NF-κB) leads to its ubiquitination and degradation, releasing NF-κB to translocate into the nucleus and induce the expression of pro-inflammatory cytokines, such as tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) [123].
For the IRF pathway, TRAF3 recruited to MAVS activates the TBK1 (TANK-binding kinase 1) and IKKε (IKK-epsilon) kinases. These kinases phosphorylate IRF3 and IRF7, transcription factors that then dimerize and translocate to the nucleus. Activated IRF3 and IRF7 drive the expression of type I interferons (IFNs), primarily IFN-α and IFN-β, which are key antiviral cytokines [124].
Another pathway activated by mitochondrial RNA involves PKR-mediated signaling. PKR (double-stranded RNA-dependent protein kinase) is a serine/threonine protein kinase initially identified as an interferon (IFN)-inducible, innate immune antiviral protein activated by dsRNA [125]. Kim et al. demonstrated that PKR senses not only viral dsRNA but also endogenous nuclear and mitochondrial dsRNA [126]. The binding of dsRNA to PKR’s dsRNA-binding domains (dsRBDs) promotes dimerization and autophosphorylation, thereby activating its kinase function [127]. Activated PKR can trigger the NF-κB signaling pathway, leading to the expression of pro-inflammatory cytokines [128]. Additionally, PKR phosphorylates the eukaryotic translation initiation factor 2α (eIF2α), inhibiting global protein synthesis to restrict viral replication [128]. Under specific conditions, PKR activation can also induce apoptosis [129].

5.2. Abnormal Mitochondrial dsRNA Accumulation Induces Immune Responses

The mitochondrial genome undergoes bidirectional transcription, generating overlapping transcripts that can form long double-stranded RNA structures [19]. Normally, the levels of mt-dsRNA are tightly regulated by the mitochondrial RNA degradosome components, such as mitochondrial RNA helicase SUV3 (hSUV3) and polynucleotide phosphorylase PNPase (PNPT1), along with its cofactor GRSF1, which have been described above [100]. However, in various pathophysiological states, this processing can be disrupted, leading to the abnormal accumulation of mtRNA. Proudfoot’s group identified PNPT1 mutation in blood leukocytes of patients. PNPT1 mutations impair PNPase function, reducing mt-dsRNA degradation and causing its accumulation. This mt-dsRNA escapes into the cytoplasm via Bax-Bak pores and is sensed by MDA5, activating the MAVS-mediated signaling pathway to trigger a type I interferon response, with elevated interferon-stimulated genes (ISGs) and markers of immune activation (Figure 2) [100]. Although SUV3 deficiency causes mt-dsRNA accumulation, it fails to trigger an immune response, even when ABT-737 (Bcl2 and Bcl-xL inhibitor) is administered [100]. The underlying mechanism for this remains unclear. Mitochondrial mRNA (mt-mRNA) is polyadenylated by mtPAP and stabilized by the LRPPRC-SLIRP complex. Studies in Drosophila revealed that, in addition to PNPase deficiency, loss of mtPAP function also promotes mt-mRNA accumulation and activates immune signaling in flies [130]. These findings indicate that aberrant accumulation of mt-dsRNA is a key factor triggering mt-RNA-mediated immune responses. Leveraging this characteristic, Sujin Kim and colleagues employed CRISPR-CAS9 screening to identify 89 mitochondrial RNA-binding proteins which, when lost, cause mt-dsRNA accumulation. Notably, they found that loss of the rRNA methyltransferase NSUN4 (NOP2/Sun RNA methyltransferase 4) promotes mt-dsRNA accumulation and immune activation through activating PKR signal pathway. Mechanistically, NSUN4 catalyzes the formation of m5C-modified RNA, which is recognized by C1q-binding protein (C1QBP). C1QBP subsequently recruits PNPT1 to the RNA for degradation, indicating that m5C marked mtRNA for degradation (Figure 2) [131].
Metabolites actively regulate mitochondrial RNA (mtRNA)-mediated immune responses. As demonstrated by Hooftman et al., LPS stimulation of macrophages induces metabolic rewiring, including an aspartate-argininosuccinate shunt driven by increased ASS1 (Argininosuccinate Synthase 1) expression [132]. This elevates cytosolic fumarate, promoting protein succination [132]. Critically, pharmacological or genetic inhibition of fumarate hydratase (FH) further increases fumarate, suppresses mitochondrial respiration, and elevates mitochondrial membrane potential. These changes reduce IL-10 and increase TNF secretion (mimicked by fumarate esters). Notably, FH inhibition uniquely triggers mtRNA release, activating RNA sensors (TLR7, RIG-I, MDA5) to induce IFN-β production, an effect also seen with endogenous FH suppression after prolonged LPS. FH suppression is observed in systemic lupus erythematosus (SLE) patient cells, suggesting its pathogenic role, indicating FH acts as a protective regulator, restraining mtRNA release to maintain appropriate cytokine and interferon responses and prevent aberrant immune activation [132].
Except for systemic lupus erythematosus (SLE), other autoimmune diseases such as Sjögren’s syndrome have also been found to have mt-dsRNA-mediated immune activation [133]. A recent study showed that the release of mt-dsRNA into the cytosol acts as a critical factor in inducing and maintaining the inflammatory phenotype of senescent cells, which is similar to mtDNA release [134,135]. Furthermore, mt-dsRNA release might be connected to viral infections and heart diseases. Recent studies have reported EMCV (encephalomyocarditis virus) infection disrupts normal mitochondrial functions, which may contribute to the release of mitochondrial dsRNA [19]. Additionally, EMCV-induced mitochondrial dysfunction and mt-dsRNA release could contribute to the tissue damage seen in infected organisms. In cases of myocarditis caused by EMCV, the leakage of mt-dsRNA into the extracellular space might recruit immune cells, exacerbating inflammation in the heart muscle. However, the potential mechanisms underlying the interplay between EMCV infection and mt-dsRNA release remain largely unelucidated and require in-depth investigation.

5.3. Mitochondrial Non-Coding RNAs Mediated Immune Responses

Ashish Dhir et al. used RNA sequencing profile to demonstrate that released mitochondrial double-stranded RNA (mt-dsRNA) covers the entire mitochondrial genome [19]. We hypothesize that mitochondrial non-coding RNAs may also act as damage-associated molecular patterns (DAMPs) capable of activating immune responses. Although there is no direct evidence supporting this hypothesis, it is plausible that mt-ncRNAs participate indirectly in immune regulation by modulating mitochondrial functions—such as oxidative phosphorylation and reactive oxygen species (ROS) production—which in turn influence inflammatory pathways.
lncFAO (fatty acid oxidation-associated lncRNA), which is nuclear-encoded mitochondria-located lncRNAs, is identified in mouse macrophages at a later time after LPS stimulation. lncFAO binds to HADHB (a subunit of the mitochondrial trifunctional enzyme, critical for β-oxidation) and increases HADHB protein levels to promote fatty acid oxidation (FAO) [136]. Fatty acid oxidation is essential for M2 macrophage polarization. By enhancing FAO, lncFAO supports M2-mediated resolution of inflammation [136].
SCAR/mc-COX2 (Steatohepatitis-associated circRNA ATP5B regulator/mt-circ-COX2), which is produced from the COX2 locus on the L-strand, is downregulated in liver fibroblasts from patients with nonalcoholic steatohepatitis (NASH) and upregulated in plasma exosomes of chronic lymphocytic leukemia (CLL) patients [137]. SCAR binds to ATP5B and inhibits the interaction between cyclophilin D (CypD, an activator of the mitochondrial permeability transition pore) and the mPTP complex. This prevents the opening of the pore and reduces the release of mitochondrial reactive oxygen species (mtROS). Deficiency in SCAR leads to accumulation of mtROS, which promotes the activation of liver fibroblasts and exacerbates metaflammation. Notably, mitochondrial delivery of SCAR using mito-NP nanoparticles has been shown to ameliorate NASH in mouse models [137].

6. Future Perspectives

Current research reveals that mt-dsRNA accumulation is a primary factor triggering RNA-mediated inflammatory responses. This aberrant RNA accumulation stems from deficiencies in the PNPT1-SUV3 complex or REXO2 function [101]. Interestingly, however, accumulation caused by SUV3 deficiency alone does not induce inflammation [19]. PNPT1 requires SUV3 as a cofactor for its RNA degradation activity and is catalytically inert when alone. PNPT1 is primarily localized to the mitochondrial matrix, although it has also been reported to degrade RNA in the intermembrane space (IMS) and cytosol [138]. Since SUV3 is exclusively expressed in the matrix, this prompts the following question: does another protein facilitate PNPT1-mediated RNA degradation within the IMS? If this protein exists, it would offer a compelling explanation for why mtRNA accumulation caused by loss of SUV3 does not induce an immune response—namely, PNPT1 may clear such RNA within the IMS.
Sujin Kim et al. reported that m5C-marked mt-rRNAs are recognized by C1QBP and subsequently degraded by PNPT1 [131]. In contrast, loss of the m5C methyltransferase NSUN4 promotes dsRNA accumulation and immune activation. This suggests that RNAs are marked for degradation. A recent study showed that pseudouridine (Ψ)-containing RNA evades immune detection by Toll-like receptors (TLR7 and TLR8) [139], indicating that RNA modification is a marker for immunity recognition. A series of key questions has emerged from these findings. Firstly, what are the specific molecular marks of RNA that play a crucial role in promoting RNA release? Do these released mt-dsRNAs carry distinct molecular signatures that promote their selective export?
Secondly, which proteins, apart from the ones already mentioned, are involved in this RNA release process? And finally, what are the underlying molecular mechanisms that govern the entire sequence of events related to RNA release? RNA typically functions in conjunction with RNA-binding proteins (RBPs). In addition to RNA modifications such as m5C and pseudouridine, which were mentioned earlier, the selective clearance or release of RNA may also be regulated by specific RBPs. These proteins could act as receptor-like molecules or adaptors that promote RNA degradation or export. So far, however, C1QBP (Complement Component 1 Q Subcomponent- Binding Protein) remains the only protein known to play such a role. We propose that other proteins with similar functions likely exist. As demonstrated by Sujin Kim et al., RNA immunoprecipitation (RIP) could serve as an effective method to isolate and identify such candidate RBPs. All these aspects are areas that demand further in-depth investigation to gain a more comprehensive understanding of the RNA-related processes in question.
How does mt-dsRNA leak from mitochondria? The mechanisms underlying mtDNA release are relatively well-characterized: mtDNA can be released into the cytosol via BAX/BAK pores, or through VDAC, the mPTP, GSDMD, and, more recently reported, p-MLKL (Mixed Lineage Kinase Domain Like Pseudokinase) pores [140,141,142,143]. Similarly, mt-dsRNA release can also occur via BAX/BAK pores and VDAC, with ANT2 recently identified as an additional released channel [19,144]. Notably, mtDNA can also be released through mitochondrial-derived vesicles (MDVs) or via fusion with multivesicular bodies (MVBs) for extracellular export [145]. Therebefore, we hypothesize that mtRNA may be released through additional mechanisms. For example, accumulated mtRNA might escape mitochondria through pores formed by MLKL or other mediators such as BSDMD, potentially triggering immune activation. Alternatively, mtRNA could be packaged into mitochondrial-derived vesicles (MDVs) and subsequently delivered to endosomes, where it engages Toll-like receptor 3 (TLR3) to initiate an immune response (Figure 2). These requires further investigation.

Funding

The work in the authors’ laboratory was supported by grants from the National Natural Science Foundation of China (82273970 to J.T., 32270768 to C.Z., 32125011 to Z.S.), the National Natural Science Foundation of Hubei (2025AFA085 to C.Z., 2024AFB765 to R.Z.). The Innovation Group Project of Hubei Province (2023AFA026 to J.T.) and the National Key R&D Program of China (2023YFC2507900 to J.T.). The Key Cultivation Project of Hubei Province for Science and Technology (2024DJA037 to J.T.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

During the preparation of this manuscript, the author used Doubao: https://www.doubao.com (accessed on 20 July 2025) and DeepSeek: https://chat.deepseek.com (accessed on 25 July 2025) for language editing, grammar correction. The author has thoroughly reviewed, edited, and validated the output and takes full responsibility for the content of this publication.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

ALKBH1AlkB Homolog 1
ASS1Argininosuccinate Synthase 1
C1QBPC1q-binding protein
DAMPsdamage-associated molecular patterns
EMCVencephalomyocarditis virus
FASTKD4FAST kinase domain-containing protein 4
GRSF1G-Rich RNA Sequence Binding Factor 1
GSDMDGasdermin D
HSPD17B10Hydroxysteroid 17-Beta Dehydrogenase 10
IFNstype I interferons
IKKphosphorylates IκB kinase
IRFInterferon regulatory factor
ISGsinterferon-stimulated genes
LRPPRCLeucine-Rich PPR Motif-Containing Protein
LSFCLeigh syndrome
MAVSMitochondrial antiviral signaling proteins
MDA5melanoma differentiation-associated gene 5
MDVsmitochondrial-derived vesicles
METTL15Methyltransferase 15
METTL17Methyltransferase Like 17
MLKLMixed Lineage Kinase Domain Like Pseudokinase
mPTPmitochondrial permeability transition pores
MRM1Mitochondrial RRNA Methyltransferase 1
MRM2Mitochondrial RRNA Methyltransferase 2
MRM3Mitochondrial RRNA Methyltransferase 3
mRNAs messenger RNAs
mtDNAmitochondrial DNA
mt-dsRNAmitochondrial double-stranded RNA
MTERF1Mitochondrial Transcription Termination Factor 1
mt-NAsmitochondrial nucleic acids
mtPAPmitochondrial poly(A) polymerase
mtRNAmitochondrial RNA
NCRnon-coding region
NF-κBNuclear Factor Kappa B Subunit 1
NSUN2NOP2/Sun RNA Methyltransferase 2
NSUN3NOP2/Sun RNA Methyltransferase 3
NSUN4NOP2/Sun RNA Methyltransferase 4
OSGEPL1O-Sialoglycoprotein Endopeptidase Like 1
PKRdouble-stranded RNA-dependent protein kinase
PNPT1PNPase, Polyribonucleotide Nucleotidyltransferase 1
POLRMTRNA polymerase, mitochondrial
PRORPProtein Only RNase P Catalytic Subunit
PUS1Pseudouridine Synthase 1
REXO2RNA Exonuclease 2
RIG-IRetinoic acid-inducible gene I
RLRsRIG-I-like receptors
RPUSD2RNA Pseudouridine Synthase Domain Containing 2
RPUSD3RNA Pseudouridine Synthase D3
RPUSD4RNA Pseudouridine Synthase D4
rRNAsribosomal RNAs
SLIRPSRA Stem-Loop Interacting RNA Binding Protein
SUV3Suv3 Like RNA Helicase
TAK1Transforming growth factor-β-activated kinase 1
TBK1TANK-binding kinase 1
TEFMTranscription elongation factor
TFAMTranscription Factor A, Mitochondrial
TFB1MTranscription Factor B1, Mitochondrial
TFB2MTranscription Factor B2, Mitochondrial
TLRstoll-like receptors
TNF-αTumor necrosis factor-α
TRAFsTumor necrosis factor receptor-associated factors
TRIT1TRNA Isopentenyltransferase 1
TRMT10CTRNA Methyltransferase 10C
TRMT5TRNA Methyltransferase 5
TRMT6TRNA Methyltransferase 6
TRMT61BTRNA Methyltransferase 61B
tRNAstransfer RNAs
TRNT1TRNA Nucleotidyl Transferase 1
TRUB2TruB Pseudouridine Synthase Family Member 2
VDACvoltage-dependent anion channel
YRDCYrdC N6-Threonylcarbamoyltransferase Domain Containing
ΨPseudouridine

References

  1. Bogenhagen, D.F. Mitochondrial DNA nucleoid structure. Biochim. Biophys. Acta-Gene Regul. Mech. 2012, 1819, 914–920. [Google Scholar] [CrossRef] [PubMed]
  2. Kakudji, E.V.; Lewis, S.C. Mitochondrial nucleoids. Curr. Biol. 2024, 34, R1067–R1068. [Google Scholar] [CrossRef] [PubMed]
  3. Sahayasheela, V.J.; Yu, Z.; Hidaka, T.; Pandian, G.N.; Sugiyama, H. Mitochondria and G-quadruplex evolution: An intertwined relationship. Trends Genet. 2023, 39, 15–30. [Google Scholar] [CrossRef]
  4. Tan, B.G.; Gustafsson, C.M.; Falkenberg, M. Mechanisms and regulation of human mitochondrial transcription. Nat. Rev. Mol. Cell Biol. 2024, 25, 119–132. [Google Scholar] [CrossRef]
  5. Hall-Younger, E.; Tait, S.W. Mitochondria and cell death signalling. Curr. Opin. Cell Biol. 2025, 94, 102510. [Google Scholar] [CrossRef]
  6. Poltorak, A. Cell death: All roads lead to mitochondria. Curr. Biol. 2022, 32, R891–R894. [Google Scholar] [CrossRef]
  7. Picard, M.; Shirihai, O.S. Mitochondrial signal transduction. Cell Metab. 2022, 34, 1620–1653. [Google Scholar] [CrossRef]
  8. Mills, E.L.; Kelly, B.; O’Neill, L.A.J. Mitochondria are the powerhouses of immunity. Nat. Immunol. 2017, 18, 488–498. [Google Scholar] [CrossRef]
  9. Gokhale, N.S.; Sam, R.K.; Somfleth, K.; Thompson, M.G.; Marciniak, D.M.; Smith, J.R.; Genoyer, E.; Eggenberger, J.; Chu, L.H.; Park, M.; et al. Cellular RNA interacts with MAVS to promote antiviral signaling. Science 2024, 386, eadl0429. [Google Scholar] [CrossRef]
  10. Xu, L.-G.; Wang, Y.-Y.; Han, K.-J.; Li, L.-Y.; Zhai, Z.; Shu, H.-B. VISA is an adapter protein required for virus-triggered IFN-β signaling. Mol. Cell. 2005, 19, 727–740. [Google Scholar] [CrossRef] [PubMed]
  11. Seth, R.B.; Sun, L.; Ea, C.-K.; Chen, Z.J. Identification and Characterization of MAVS, a Mitochondrial Antiviral Signaling Protein that Activates NF-κB and IRF3. Cell 2005, 122, 669–682. [Google Scholar] [CrossRef] [PubMed]
  12. Meylan, E.; Curran, J.; Hofmann, K.; Moradpour, D.; Binder, M.; Bartenschlager, R.; Tschopp, J. Cardif is an adaptor protein in the RIG-I antiviral pathway and is targeted by hepatitis C virus. Nature 2005, 437, 1167–1172. [Google Scholar] [CrossRef]
  13. Kawai, T.; Takahashi, K.; Sato, S.; Coban, C.; Kumar, H.; Kato, H.; Ishii, K.J.; Takeuchi, O.; Akira, S. IPS-1, an adaptor triggering RIG-I- and Mda5-mediated type I interferon induction. Nat. Immunol. 2005, 6, 981–988. [Google Scholar] [CrossRef] [PubMed]
  14. Hu, M.M.; Shu, H.B. Mitochondrial DNA-triggered innate immune response: Mechanisms and diseases. Cell. Mol. Immunol. 2023, 20, 1403–1412. [Google Scholar] [CrossRef]
  15. Newman, L.E.; Novak, S.W.; Rojas, G.R.; Tadepalle, N.; Schiavon, C.R.; Grotjahn, D.A.; Towers, C.G.; Tremblay, M.E.; Donnelly, M.P.; Ghosh, S.; et al. Mitochondrial DNA replication stress triggers a pro-inflammatory endosomal pathway of nucleoid disposal. Nat. Cell Biol. 2024, 26, 194–206. [Google Scholar] [CrossRef]
  16. McArthur, K.; Whitehead, L.W.; Heddleston, J.M.; Li, L.; Padman, B.S.; Oorschot, V.; Geoghegan, N.D.; Chappaz, S.; Davidson, S.; Chin, H.S.; et al. BAK/BAX macropores facilitate mitochondrial herniation and mtDNA efflux during apoptosis. Science 2018, 359, 883. [Google Scholar] [CrossRef]
  17. Yu, C.H.; Davidson, S.; Harapas, C.R.; Hilton, J.B.; Mlodzianoski, M.J.; Laohamonthonkul, P.; Louis, C.; Low, R.R.J.; Moecking, J.; De Nardo, D.; et al. TDP-43 Triggers Mitochondrial DNA Release via mPTP to Activate cGAS/STING in ALS. Cell 2020, 183, 636. [Google Scholar] [CrossRef]
  18. Xian, H.X.; Watari, K.; Sanchez-Lopez, E.; Offenberger, J.; Onyuru, J.; Sampath, H.; Ying, W.; Hoffman, H.M.; Shadel, G.S.; Karin, M. Oxidized DNA fragments exit mitochondria via mPTP- and VDAC-dependent channels to activate NLRP3 inflammasome and interferon signaling. Immunity 2022, 55, 1370. [Google Scholar] [CrossRef]
  19. Dhir, A.; Dhir, S.; Borowski, L.S.; Jimenez, L.; Teitell, M.; Rötig, A.; Crow, Y.J.; Rice, G.I.; Duffy, D.; Tamby, C.; et al. Mitochondrial double-stranded RNA triggers antiviral signalling in humans. Nature 2018, 560, 238. [Google Scholar] [CrossRef]
  20. Krieger, M.R.; Abrahamian, M.; He, K.L.; Atamdede, S.; Hakimjavadi, H.; Momcilovic, M.; Ostrow, D.; Maggo, S.D.; Tsang, Y.P.; Gai, X.; et al. Trafficking of mitochondrial double-stranded RNA from mitochondria to the cytosol. Life Sci. Alliance 2024, 7, e202302396. [Google Scholar] [CrossRef] [PubMed]
  21. Morozov, Y.I.; Agaronyan, K.; Cheung, A.C.M.; Anikin, M.; Cramer, P.; Temiakov, D. A novel intermediate in transcription initiation by human mitochondrial RNA polymerase. Nucleic Acids Res. 2014, 42, 3884–3893. [Google Scholar] [CrossRef]
  22. Hillen, H.S.; Morozov, Y.I.; Sarfallah, A.; Temiakov, D.; Cramer, P. Structural Basis of Mitochondrial Transcription Initiation. Cell 2017, 171, 1072. [Google Scholar] [CrossRef]
  23. Song, Y.; Wang, W.J.; Wang, B.B.; Shi, Q.W. The Protective Mechanism of TFAM on Mitochondrial DNA and its Role in Neurodegenerative Diseases. Mol. Neurobiol. 2024, 61, 4381–4390. [Google Scholar] [PubMed]
  24. Rubio-Cosials, A.; Sidow, J.F.; Jiménez-Menéndez, N.; Fernández-Millán, P.; Montoya, J.; Jacobs, H.T.; Coll, M.; Bernadó, P.; Solà, M. Human mitochondrial transcription factor A induces a U-turn structure in the light strand promoter. Nat. Struct. Mol. Biol. 2012, 19, 365. [Google Scholar] [CrossRef]
  25. Malarkey, C.S.; Bestwick, M.; Kuhlwilm, J.E.; Shadel, G.S.; Churchill, M.E.A. Transcriptional activation by mitochondrial transcription factor A involves preferential distortion of promoter DNA. Nucleic Acids Res. 2012, 40, 614–624. [Google Scholar] [CrossRef] [PubMed]
  26. Morozov, Y.I.; Parshin, A.V.; Agaronyan, K.; Cheung, A.C.M.; Anikin, M.; Cramer, P.; Temiakov, D. A model for transcription initiation in human mitochondria. Nucleic Acids Res. 2015, 43, 3726–3735. [Google Scholar] [CrossRef]
  27. Hillen, H.S.; Temiakov, D.; Cramer, P. Structural basis of mitochondrial transcription. Nat. Struct. Mol. Biol. 2018, 25, 754–765. [Google Scholar] [CrossRef]
  28. Bogenhagen, D.F.; Martin, D.W.; Koller, A. Initial Steps in RNA Processing and Ribosome Assembly Occur at Mitochondrial DNA Nucleoids. Cell Metab. 2014, 19, 618–629. [Google Scholar] [CrossRef]
  29. Terzioglu, M.; Ruzzenente, B.; Harmel, J.; Mourier, A.; Jemt, E.; López, M.D.; Kukat, C.; Stewart, J.B.; Wibom, R.; Meharg, C.; et al. MTERF1 Binds mtDNA to Prevent Transcriptional Interference at the Light-Strand Promoter but Is Dispensable for rRNA Gene Transcription Regulation. Cell Metab. 2013, 17, 618–626. [Google Scholar] [CrossRef]
  30. Yakubovskaya, E.; Mejia, E.; Byrnes, J.; Hambardjieva, E.; Garcia-Diaz, M. Helix Unwinding and Base Flipping Enable Human MTERF1 to Terminate Mitochondrial Transcription. Cell 2010, 141, 982–993. [Google Scholar] [CrossRef] [PubMed]
  31. Gustafsson, C.M.; Larsson, N.G. MTERF1 Gives mtDNA an Unusual Twist. Cell Metab. 2010, 12, 3–4. [Google Scholar] [CrossRef]
  32. Byrnes, J.; Hauser, K.; Norona, L.; Mejia, E.; Simmerling, C.; Garcia-Diaz, M. Base Flipping by MTERF1 Can Accommodate Multiple Conformations and Occurs in a Stepwise Fashion. J. Mol. Biol. 2016, 428, 2542–2556. [Google Scholar] [CrossRef] [PubMed]
  33. Martin, M.; Cho, J.Y.; Cesare, A.J.; Griffith, J.D.; Attardi, G. Termination factor-mediated DNA loop between termination and initiation sites drives mitochondrial rRNA synthesis. Cell 2005, 123, 1227–1240. [Google Scholar] [CrossRef]
  34. Jedynak-Slyvka, M.; Jabczynska, A.; Szczesny, R.J. Human Mitochondrial RNA Processing and Modifications: Overview. Int. J. Mol. Sci. 2021, 22, 7999. [Google Scholar] [CrossRef]
  35. Barchiesi, A.; Vascotto, C. Transcription, Processing, and Decay of Mitochondrial RNA in Health and Disease. Int. J. Mol. Sci. 2019, 20, 2221. [Google Scholar] [CrossRef]
  36. Burger, G.; Citterich, M.H.; Nelson, M.A.; Werner, S.; Macino, G. Rna Processing in Neurospora-Crassa Mitochondria—Transfer-Rnas Punctuate a Large Precursor Transcript. EMBO J. 1985, 4, 197–204. [Google Scholar] [CrossRef]
  37. Ojala, D.; Montoya, J.; Attardi, G. tRNA punctuation model of RNA processing in human mitochondria. Nature 1981, 290, 470–474. [Google Scholar] [CrossRef] [PubMed]
  38. Holzmann, J.; Frank, P.; Löffler, E.; Bennett, K.L.; Gerner, C.; Rossmanith, W. RNase P without RNA: Identification and Functional Reconstitution of the Human Mitochondrial tRNA Processing Enzyme. Cell 2008, 135, 462–474. [Google Scholar] [CrossRef] [PubMed]
  39. Redko, Y.; de la Sierra-Gallay, I.L.; Condon, C. When all’s zed and done: The structure and function of RNase Z in prokaryotes. Nat. Rev. Microbiol. 2007, 5, 278–286. [Google Scholar] [CrossRef]
  40. de la Sierra-Gallay, I.L.; Pellegrini, O.; Condon, C. Structural basis for substrate binding, cleavage and allostery in the tRNA maturase RNase Z. Nature 2005, 433, 657–661. [Google Scholar] [CrossRef]
  41. Bhatta, A.; Dienemann, C.; Cramer, P.; Hillen, H.S. Structural basis of RNA processing by human mitochondrial RNase P. Nat. Struct. Mol. Biol. 2021, 28, 713. [Google Scholar] [CrossRef]
  42. Evans, D.; Marquez, S.M.; Pace, N.R. RNase P: Interface of the RNA and protein worlds. Trends Biochem. Sci. 2006, 31, 333–341. [Google Scholar] [CrossRef]
  43. Esakova, O.; Krasilnikov, A.S. Of proteins and RNA: The RNase P/MRP family. RNA 2010, 16, 1725–1747. [Google Scholar] [CrossRef]
  44. Reinhard, L.; Sridhara, S.; Hällberg, B.M. Structure of the nuclease subunit of human mitochondrial RNase P. Nucleic Acids Res. 2015, 43, 5664–5672. [Google Scholar] [CrossRef] [PubMed]
  45. Meynier, V.; Hardwick, S.W.; Catala, M.; Roske, J.J.; Oerum, S.; Chirgadze, D.Y.; Barraud, P.; Yue, W.W.; Luisi, B.; Tisné, C. Structural basis for human mitochondrial tRNA maturation. Nat. Commun. 2024, 15, 4683. [Google Scholar] [CrossRef]
  46. Reinhard, L.; Sridhara, S.; Hällberg, B.M. The MRPP1/MRPP2 complex is a tRNA-maturation platform in human mitochondria. Nucleic Acids Res. 2017, 45, 12469–12480. [Google Scholar] [CrossRef]
  47. Ohkubo, A.; Van Haute, L.S.; Rudler, D.; Stentenbach, M.S.; Steiner, F.; Rackham, O.S.; Minczuk, M.; Filipovska, A.S.; Martinou, J.C. The FASTK family proteins fine-tune mitochondrial RNA processing. PLoS Genet. 2021, 17, e1009873. [Google Scholar] [CrossRef]
  48. Rackham, O.; Shearwood, A.-M.J.; Mercer, T.R.; Davies, S.M.; Mattick, J.S.; Filipovska, A. Long noncoding RNAs are generated from the mitochondrial genome and regulated by nuclear-encoded proteins. RNA. 2011, 17, 2085–2093. [Google Scholar] [CrossRef]
  49. Liu, X.; Wang, X.L.; Li, J.X.; Hu, S.S.; Deng, Y.Q.; Yin, H.; Bao, X.C.; Zhang, Q.C.; Wang, G.; Wang, B.L.; et al. Identification of mecciRNAs and their roles in the mitochondrial entry of proteins. Sci. China Life Sci. 2020, 63, 1429–1449. [Google Scholar] [CrossRef] [PubMed]
  50. Ren, B.; Guan, M.X.; Zhou, T.; Cai, X.; Shan, G. Emerging functions of mitochondria-encoded noncoding RNAs. Trends Genet. 2023, 39, 125–139. [Google Scholar] [CrossRef] [PubMed]
  51. Sun, W.H.; Lu, Y.J.; Zhang, H.; Zhang, J.; Fang, X.Y.; Wang, J.X.; Li, M.Y. Mitochondrial Non-Coding RNAs Are Potential Mediators of Mitochondrial Homeostasis. Biomolecules 2022, 12, 1863. [Google Scholar] [CrossRef] [PubMed]
  52. Zhu, X.F.; Xie, X.; Das, H.; Tan, B.G.; Shi, Y.H.; Al-Behadili, A.; Peter, B.; Motori, E.; Valenzuela, S.; Posse, V.; et al. Non-coding 7S RNA inhibits transcription via mitochondrial RNA polymerase dimerization. Cell 2022, 185, 2309. [Google Scholar] [CrossRef]
  53. Rorbach, J.; Minczuk, M. The post-transcriptional life of mammalian mitochondrial RNA. Biochem. J. 2012, 444, 357–373. [Google Scholar] [CrossRef]
  54. Mercer, T.R.; Neph, S.; Dinger, M.E.; Crawford, J.; Smith, M.A.; Shearwood, A.M.J.; Haugen, E.; Bracken, C.P.; Rackham, O.; Stamatoyannopoulos, J.A.; et al. The Human Mitochondrial Transcriptome. Cell 2011, 146, 645–658. [Google Scholar] [CrossRef]
  55. Temperley, R.J.; Seneca, S.H.; Tonska, K.; Bartnik, E.; Bindoff, L.A.; Lightowlers, R.N.; Chrzanowska-Lightowlers, Z.M.A. Investigation of a pathogenic mtDNA microdeletion reveals a translation-dependent deadenylation decay pathway in human mitochondria. Hum. Mol. Genet. 2003, 12, 2341–2348. [Google Scholar] [CrossRef]
  56. Bratic, A.; Clemente, P.; Calvo-Garrido, J.; Maffezzini, C.; Felser, A.; Wibom, R.; Wedell, A.; Freyer, C.; Wredenberg, A. Polyadenylation of mitochondrial transcripts is exclusively performed by mitochondrial poly(A) polymerase. Mitochondrion 2016, 31, 92. [Google Scholar]
  57. Chang, J.H.; Tong, L. Mitochondrial poly(A) polymerase and polyadenylation. Biochim. Biophys. Acta-Gene Regul. Mech. 2012, 1819, 992–997. [Google Scholar] [CrossRef] [PubMed]
  58. Sachs, A. The role of poly(A) in the translation and stability of mRNA. Curr. Opin. Cell Biol. 1990, 2, 1092–1098. [Google Scholar] [CrossRef]
  59. Mishra, V.K.; Wegwitz, F.; Kosinsky, R.L.; Sen, M.; Baumgartner, R.; Wulff, T.; Siveke, J.T.; Schildhaus, H.U.; Najafova, Z.; Kari, V.; et al. Histone deacetylase class-I inhibition promotes epithelial gene expression in pancreatic cancer cells in a BRD4-and MYC-dependent manner. Nucleic Acids Res. 2017, 45, 6334–6349. [Google Scholar] [CrossRef]
  60. Wilson, W.C.; Hornig-Do, H.T.; Bruni, F.; Chang, J.H.; Jourdain, A.A.; Martinou, J.C.; Falkenberg, M.; Spåhr, H.; Larsson, N.G.; Lewis, R.J.; et al. A human mitochondrial poly(A) polymerase mutation reveals the complexities of post-transcriptional mitochondrial gene expression. Hum. Mol. Genet. 2014, 23, 6345–6355. [Google Scholar] [CrossRef]
  61. Crosby, A.H.; Patel, H.; Chioza, B.A.; Proukakis, C.; Gurtz, K.; Patton, M.A.; Sharifi, R.; Harlalka, G.; Simpson, M.A.; Dick, K.; et al. Defective Mitochondrial mRNA Maturation Is Associated with Spastic Ataxia. Am. J. Hum. Genet. 2010, 87, 655–660. [Google Scholar] [CrossRef]
  62. Siira, S.J.; Spåhr, H.; Shearwood, A.M.J.; Ruzzenente, B.; Larsson, N.G.; Rackham, O.; Filipovska, A. LRPPRC-mediated folding of the mitochondrial transcriptome. Nat. Commun. 2017, 8, 1532. [Google Scholar] [CrossRef]
  63. Rubalcava-Gracia, D.; Bubb, K.; Levander, F.; Burr, S.P.; August, A.; Chinnery, P.F.; Koolmeister, C.; Larsson, N.G. LRPPRC and SLIRP synergize to maintain sufficient and orderly mammalian mitochondrial translation. Nucleic Acids Res. 2024, 52, 11266–11282. [Google Scholar] [CrossRef]
  64. Lagouge, M.; Mourier, A.; Lee, H.J.; Spåhr, H.; Wai, T.; Kukat, C.; Ramos, E.S.; Motori, E.; Busch, J.D.; Siira, S.; et al. SLIRP Regulates the Rate of Mitochondrial Protein Synthesis and Protects LRPPRC from Degradation. PLoS Genet. 2015, 11, e1005423. [Google Scholar] [CrossRef] [PubMed]
  65. Singh, V.; Moran, J.C.; Itoh, Y.; Soto, I.C.; Fontanesi, F.; Couvillion, M.; Huynen, M.A.; Churchman, L.S.; Barrientos, A.; Amunts, A. Structural basis of LRPPRC-SLIRP-dependent translation by the mitoribosome. Nat. Struct. Mol. Biol. 2024, 31, 1838–1847. [Google Scholar] [CrossRef] [PubMed]
  66. Ruzzenente, B.; Metodiev, M.D.; Wredenberg, A.; Bratic, A.; Park, C.B.; Cámara, Y.; Milenkovic, D.; Zickermann, V.; Wibom, R.; Hultenby, K.; et al. LRPPRC is necessary for polyadenylation and coordination of translation of mitochondrial mRNAs. EMBO J. 2012, 31, 443–456. [Google Scholar] [CrossRef] [PubMed]
  67. Huang, H.L.; Weng, H.Y.; Chen, J.J. m6A Modification in Coding and Non-coding RNAs: Roles and Therapeutic Implications in Cancer. Cancer Cell 2020, 37, 270–288. [Google Scholar]
  68. Karijolich, J.; Yi, C.Q.; Yu, Y.T. Transcriptome-wide dynamics of RNA pseudouridylation. Nat. Rev. Mol. Cell Biol. 2015, 16, 581–585. [Google Scholar] [CrossRef]
  69. Li, X.Y.; Xiong, X.S.; Zhang, M.L.; Wang, K.; Chen, Y.; Zhou, J.; Mao, Y.H.; Lv, J.; Yi, D.Y.; Chen, X.W.; et al. Base-Resolution Mapping Reveals Distinct m6A Methylome in Nuclear- and Mitochondrial-Encoded Transcripts. Mol. Cell 2017, 68, 993. [Google Scholar] [CrossRef]
  70. Antonicka, H.; Choquet, K.; Lin, Z.Y.; Gingras, A.C.; Kleinman, C.L.; Shoubridge, E.A. A pseudouridine synthase module is essential for mitochondrial protein synthesis and cell viability. EMBO Rep. 2017, 18, 28–38. [Google Scholar] [CrossRef]
  71. Hao, Z.Y.; Wu, T.; Cui, X.L.; Zhu, P.P.; Tan, C.P.; Dou, X.Y.; Hsu, K.W.; Lin, Y.T.; Peng, P.H.; Zhang, L.S.; et al. -Deoxyadenosine Methylation in Mammalian Mitochondrial DNA. Mol. Cell 2020, 78, 382. [Google Scholar] [CrossRef]
  72. Nagaike, T.; Suzuki, T.; Katoh, T.; Ueda, T. Human mitochondrial mRNAs are stabilized with polyadenylation regulated by mitochondria-specific poly(A) polymerase and polynucleotide phosphorylase. J. Biol. Chem. 2005, 280, 19721–19727. [Google Scholar] [CrossRef]
  73. Fatica, T.; Naas, T.; Liwak, U.; Slaa, H.; Souaid, M.; Frangione, B.; Kattini, R.; Gaudreau-Lapierre, A.; Trinkle-Mulcahy, L.; Chakraborty, P.; et al. TRNT-1 Deficiency Is Associated with Loss of tRNA Integrity and Imbalance of Distinct Proteins. Genes 2023, 14, 1043. [Google Scholar] [CrossRef]
  74. Chakraborty, P.K.; Schmitz-Abe, K.; Kennedy, E.K.; Mamady, H.; Naas, T.; Durie, D.; Campagna, D.R.; Lau, A.; Sendamarai, A.K.; Wiseman, D.H.; et al. Mutations in cause congenital sideroblastic anemia with immunodeficiency, fevers, and developmental delay (SIFD). Blood 2014, 124, 2867–2871. [Google Scholar] [CrossRef] [PubMed]
  75. Vilardo, E.; Nachbagauer, C.; Buzet, A.; Taschner, A.; Holzmann, J.; Rossmanith, W. A subcomplex of human mitochondrial RNase P is a bifunctional methyltransferase—Extensive moonlighting in mitochondrial tRNA biogenesis. Nucleic Acids Res. 2012, 40, 11583–11593. [Google Scholar] [CrossRef] [PubMed]
  76. Falk, M.J.; Gai, X.W.; Shigematsu, M.; Vilardo, E.; Takase, R.; McCormick, E.; Christian, T.; Place, E.; Pierce, E.A.; Consugar, M.; et al. A novel mutation impairing the activities of the mitochondrial RNase P complex causes X-linked intractable epilepsy and neurodevelopmental regression. RNA Biol. 2016, 13, 477–485. [Google Scholar] [CrossRef] [PubMed]
  77. Van Haute, L.; Lee, S.Y.; McCann, B.J.; Powell, C.A.; Bansal, D.; Vasiliauskaite, L.; Garone, C.; Shin, S.; Kim, J.S.; Frye, M.; et al. NSUN2 introduces 5-methylcytosines in mammalian mitochondrial tRNAs. Nucleic Acids Res. 2019, 47, 8720–8733. [Google Scholar] [CrossRef]
  78. Bohnsack, M.T.; Sloan, K.E. The mitochondrial epitranscriptome: The roles of RNA modifications in mitochondrial translation and human disease. Cell. Mol. Life Sci. 2018, 75, 241–260. [Google Scholar] [CrossRef]
  79. Zhao, X.S.; Patton, J.R.; Davis, S.L.; Florence, B.; Ames, S.J.; Spanjaard, R.A. Regulation of nuclear receptor activity by a pseudouridine synthase through posttranscriptional modification of steroid receptor RNA activator. Mol. Cell 2004, 15, 549–558. [Google Scholar] [CrossRef]
  80. Li, X.Y.; Ma, S.Q.; Yi, C.Q. Pseudouridine: The fifth RNA nucleotide with renewed interests. Curr. Opin. Chem. Biol. 2016, 33, 108–116. [Google Scholar] [CrossRef]
  81. Yuan, W.B.; Zhang, R.; Lyu, H.; Xiao, S.; Guo, D.; Zhang, Q.; Ali, D.W.; Michalak, M.; Chen, X.Z.; Zhou, C.; et al. Dysregulation of tRNA methylation in cancer: Mechanisms and targeting therapeutic strategies. Cell Death Discov. 2024, 10, 327. [Google Scholar] [CrossRef]
  82. Wu, Z.J.; Zhou, R.X.; Li, B.Z.; Cao, M.Y.; Wang, W.L.; Li, X.Y. Methylation modifications in tRNA and associated disorders: Current research and potential therapeutic targets. Cell Prolif. 2024, 57, e13692. [Google Scholar] [CrossRef]
  83. Trixl, L.; Amort, T.; Wille, A.; Zinni, M.; Ebner, S.; Hechenberger, C.; Eichin, F.; Gabriel, H.; Schoberleitner, I.; Huang, A.M.; et al. RNA cytosine methyltransferase Nsun3 regulates embryonic stem cell differentiation by promoting mitochondrial activity. Cell Mol. Life Sci. 2018, 75, 1483–1497. [Google Scholar] [CrossRef]
  84. Powel, C.A.; Kopajtich, R.; D’Souza, A.R.; Rorbach, J.; Kremer, L.S.; Husain, R.A.; Dallabona, C.; Donnini, C.; Alston, C.L.; Griffin, H.; et al. Mutations Cause a Defect in Post-transcriptional Modification of Mitochondrial tRNA Associated with Multiple Respiratory-Chain Deficiencies. Am. J. Hum. Genet. 2015, 97, 319–328. [Google Scholar] [CrossRef]
  85. Yarham, J.W.; Lamichhane, T.N.; Pyle, A.; Mattijssen, S.; Baruffini, E.; Bruni, F.; Donnini, C.; Vassilev, A.; He, L.P.; Blakely, E.L.; et al. Defective i6A37 Modification of Mitochondrial and Cytosolic tRNAs Results from Pathogenic Mutations in TRIT1 and Its Substrate tRNA. PLoS Genet. 2014, 10, e1004424. [Google Scholar] [CrossRef]
  86. Zhou, J.B.; Wang, Y.; Zeng, Q.Y.; Meng, S.X.; Wang, E.D.; Zhou, X.L. Molecular basis for tA modification in human mitochondria. Nucleic Acids Res. 2020, 48, 3181–3194. [Google Scholar] [CrossRef]
  87. Lee, K.W.; Bogenhagen, D.F. Assignment of 2′-O-Methyltransferases to Modification Sites on the Mammalian Mitochondrial Large Subunit 16 S Ribosomal RNA (rRNA). J. Biol. Chem. 2014, 289, 24936–24942. [Google Scholar] [CrossRef]
  88. Rorbach, J.; Boesch, P.; Gammage, P.A.; Nicholls, T.J.J.; Pearce, S.F.; Patel, D.; Hauser, A.; Perocchi, F.; Minczuk, M. MRM2 and MRM3 are involved in biogenesis of the large subunit of the mitochondrial ribosome. Mol. Biol. Cell 2014, 25, 2542–2555. [Google Scholar] [CrossRef] [PubMed]
  89. Bar-Yaacov, D.; Frumkin, I.; Yashiro, Y.; Chujo, T.; Ishigami, Y.; Chemla, Y.; Blumberg, A.; Schlesinger, O.; Bieri, P.; Greber, B.; et al. Mitochondrial 16S rRNA Is Methylated by tRNA Methyltransferase TRMT61B in All Vertebrates. PLoS Biol. 2016, 14, e1002557. [Google Scholar] [CrossRef] [PubMed]
  90. Zaganelli, S.; Rebelo-Guiomar, P.; Maundrell, K.; Rozanska, A.; Pierredon, S.; Powell, C.A.; Jourdain, A.A.; Hulo, N.; Lightowlers, R.N.; Chrzanowska-Lightowlers, Z.M.; et al. The Pseudouridine Synthase RPUSD4 Is an Essential Component of Mitochondrial RNA Granules. J. Biol. Chem. 2017, 292, 4519–4532. [Google Scholar] [CrossRef] [PubMed]
  91. Van Haute, L.; Hendrick, A.G.; D’Souza, A.R.; Powell, C.A.; Rebelo-Guiomar, P.; Harbour, M.E.; Ding, S.J.; Fearnley, I.M.; Andrews, B.; Minczuk, M. METTL15 introduces N4-methylcytidine into human mitochondrial 12S rRNA and is required for mitoribosome biogenesis. Nucleic Acids Res. 2019, 47, 10267–10281. [Google Scholar] [CrossRef] [PubMed]
  92. Metodiev, M.D.; Spåhr, H.; Polosa, P.L.; Meharg, C.; Becker, C.; Altmueller, J.; Habermann, B.; Larsson, N.G.; Ruzzenente, B. NSUN4 Is a Dual Function Mitochondrial Protein Required for Both Methylation of 12S rRNA and Coordination of Mitoribosomal Assembly. PLoS Genet. 2014, 10, e1004110. [Google Scholar] [CrossRef]
  93. Shi, Z.N.; Xu, S.Y.; Xing, S.H.; Yao, K.; Zhang, L.; Xue, L.X.; Zhou, P.; Wang, M.; Yan, G.Q.; Yang, P.Y.; et al. Mettl17, a regulator of mitochondrial ribosomal RNA modifications, is required for the translation of mitochondrial coding genes. FASEB J. 2019, 33, 13040–13050. [Google Scholar] [CrossRef] [PubMed]
  94. Arroyo, J.D.; Jourdain, A.A.; Calvo, S.E.; Ballarano, C.A.; Doench, J.G.; Root, D.E.; Mootha, V.K. A Genome-wide CRISPR Death Screen Identifies Genes Essential for Oxidative Phosphorylation. Cell Metab. 2016, 24, 875–885. [Google Scholar] [CrossRef] [PubMed]
  95. Li, H.; Yu, K.L.; Hu, H.L.; Zhang, X.D.; Zeng, S.Y.; Li, J.W.; Dong, X.N.; Deng, X.S.; Zhang, J.H.; Zhang, Y.Y. METTL17 coordinates ferroptosis and tumorigenesis by regulating mitochondrial translation in colorectal cancer. Redox Biol. 2024, 71, 103087. [Google Scholar] [CrossRef]
  96. Chen, H.; Shi, Z.N.; Guo, J.J.; Chang, K.J.; Chen, Q.Q.; Yao, C.H.; Haigis, M.C.; Shi, Y. The human mitochondrial 12S rRNA m5C methyltransferase METTL15 is required for mitochondrial function. J. Biol. Chem. 2020, 295, 8505–8513. [Google Scholar] [CrossRef]
  97. Seidel-Rogol, B.L.; McCulloch, V.; Shadel, G.S. Human mitochondrial transcription factor B1 methylates ribosomal RNA at a conserved stem-loop. Nat. Genet. 2003, 33, 23–24. [Google Scholar] [CrossRef]
  98. Rozanska, A.; Richter-Dennerlein, R.; Rorbach, J.; Gao, F.; Lewis, R.J.; Chrzanowska-Lightowlers, Z.M.; Lightowlers, R.N. The human RNA-binding protein RBFA promotes the maturation of the mitochondrial ribosome. Biochem. J. 2017, 474, 2145–2158. [Google Scholar] [CrossRef]
  99. Borowski, L.S.; Dziembowski, A.; Hejnowicz, M.S.; Stepien, P.P.; Szczesny, R.J. Human mitochondrial RNA decay mediated by PNPase–hSuv3 complex takes place in distinct foci. Nucleic Acids Res. 2012, 41, 1223–1240. [Google Scholar] [CrossRef]
  100. Pietras, Z.; Wojcik, M.A.; Borowski, L.S.; Szewczyk, M.; Kulinski, T.M.; Cysewski, D.; Stepien, P.P.; Dziembowski, A.; Szczesny, R.J. Controlling the mitochondrial antisense—Role of the SUV3-PNPase complex and its co-factor GRSF1 in mitochondrial RNA surveillance. Mol. Cell. Oncol. 2018, 5, e1516452. [Google Scholar] [CrossRef]
  101. Szewczyk, M.; Malik, D.; Borowski, L.S.; Czarnomska, S.D.; Kotrys, A.V.; Klosowska-Kosicka, K.; Nowotny, M.; Szczesny, R.J. Human REXO2 controls short mitochondrial RNAs generated by mtRNA processing and decay machinery to prevent accumulation of double-stranded RNA. Nucleic Acids Res. 2020, 48, 5572–5590. [Google Scholar] [CrossRef]
  102. Sasarman, F.; Brunel-Guitton, C.; Antonicka, H.; Wai, T.; Shoubridge, E.A.; Consortium, L. LRPPRC and SLIRP Interact in a Ribonucleoprotein Complex That Regulates Posttranscriptional Gene Expression in Mitochondria. Mol. Biol. Cell 2010, 21, 1315–1323. [Google Scholar] [CrossRef]
  103. Bruni, F.; Proctor-Kent, Y.; Lightowlers, R.N.; Chrzanowska-Lightowlers, Z.M. Messenger RNA delivery to mitoribosomes—Hints from a bacterial toxin. FEBS J. 2021, 288, 437–451. [Google Scholar] [CrossRef]
  104. Wang, G.; Chen, H.W.; Oktay, Y.; Zhang, J.; Allen, E.L.; Smith, G.M.; Fan, K.C.; Hong, J.S.; French, S.W.; McCaffery, J.M.; et al. PNPASE Regulates RNA Import into Mitochondria. Cell 2010, 142, 456–467. [Google Scholar] [CrossRef]
  105. von Ameln, S.; Wang, G.; Boulouiz, R.; Rutherford, M.A.; Smith, G.M.; Li, Y.; Pogoda, H.M.; Nürnberg, G.; Stiller, B.; Volk, A.E.; et al. A Mutation in, Encoding Mitochondrial-RNA-Import Protein PNPase, Causes Hereditary Hearing Loss. Am. J. Hum. Genet. 2012, 91, 919–927. [Google Scholar] [CrossRef]
  106. Vedrenne, V.; Gowher, A.; De Lonlay, P.; Nitschke, P.; Serre, V.; Boddaert, N.; Altuzarra, C.; Mager-Heckel, A.M.; Chretien, F.; Entelis, N.; et al. Mutation in, which Encodes a Polyribonucleotide Nucleotidyltransferase, Impairs RNA Import into Mitochondria and Causes Respiratory-Chain Deficiency. Am. J. Hum. Genet. 2012, 91, 912–918. [Google Scholar] [CrossRef] [PubMed]
  107. Tang, J.X.; Li, J.; Qin, S.Y.; Xiao, Y.; Liu, J.X.; Chen, X.; Zhang, Y.Y. The Biological Role of LRPPRC in Human Cancers. Cancer Control 2025, 32, 10732748251353077. [Google Scholar] [CrossRef] [PubMed]
  108. Xue, Q.Q.; Wang, W.X.; Liu, J.; Wang, D.C.; Zhang, T.Y.; Shen, T.T.; Liu, X.S.; Wang, X.J.; Shao, X.Y.; Zhou, W.; et al. LRPPRC confers enhanced oxidative phosphorylation metabolism in triple-negative breast cancer and represents a therapeutic target. J. Transl. Med. 2025, 23, 372. [Google Scholar] [CrossRef] [PubMed]
  109. Chan, Y.K.; Gack, M.U. Viral evasion of intracellular DNA and RNA sensing. Nat. Rev. Microbiol. 2016, 14, 360–373. [Google Scholar] [CrossRef]
  110. Takeuchi, O.; Akira, S. Pattern Recognition Receptors and Inflammation. Cell 2010, 140, 805–820. [Google Scholar] [CrossRef]
  111. Kawai, T.; Akira, S. The role of pattern-recognition receptors in innate immunity: Update on Toll-like receptors. Nat. Immunol. 2010, 11, 373–384. [Google Scholar] [CrossRef]
  112. Loo, Y.M.; Gale, M. Immune Signaling by RIG-I-like Receptors. Immunity 2011, 34, 680–692. [Google Scholar] [CrossRef]
  113. Goubau, D.; Schlee, M.; Deddouche, S.; Pruijssers, A.J.; Zillinger, T.; Goldeck, M.; Schuberth, C.; Van der Veen, A.G.; Fujimura, T.; Rehwinkel, J.; et al. Antiviral immunity via RIG-I-mediated recognition of RNA bearing 5′-diphosphates. Nature 2014, 514, 372. [Google Scholar] [CrossRef] [PubMed]
  114. Wu, B.; Peisley, A.; Richards, C.; Yao, H.; Zeng, X.H.; Lin, C.; Chu, F.X.; Walz, T.; Hur, S. Structural Basis for dsRNA Recognition, Filament Formation, and Antiviral Signal Activation by MDA5. Cell 2013, 152, 276–289. [Google Scholar] [CrossRef]
  115. Doyle, S.E.; Vaidya, S.A.; O’Connell, R.; Dadgostar, H.; Dempsey, P.W.; Wu, T.T.; Rao, G.; Sun, R.; Haberland, M.E.; Modlin, R.L.; et al. IRF3 mediates a TLR3/TLR4-specific antiviral gene program. Immunity 2002, 17, 251–263. [Google Scholar] [CrossRef] [PubMed]
  116. Krieg, A.M.; Vollmer, J. Toll-like receptors 7, 8, and 9: Linking innate immunity to autoimmunity. Immunol. Rev. 2007, 220, 251–269. [Google Scholar] [CrossRef]
  117. Hou, F.J.; Sun, L.J.; Zheng, H.; Skaug, B.; Jiang, Q.X.; Chen, Z.J.J. MAVS Forms Functional Prion-like Aggregates to Activate and Propagate Antiviral Innate Immune Response. Cell 2011, 146, 448–461. [Google Scholar] [CrossRef]
  118. Wu, B.; Peisley, A.; Tetrault, D.; Li, Z.L.; Egelman, E.H.; Magor, K.E.; Walz, T.; Penczek, P.A.; Hur, S. Molecular Imprinting as a Signal-Activation Mechanism of the Viral RNA Sensor RIG-I. Mol. Cell 2014, 55, 511–523. [Google Scholar] [CrossRef]
  119. Paz, S.; Vilasco, M.; Werden, S.J.; Arguello, M.; Joseph-Pillai, D.; Zhao, T.; Nguyen, T.L.A.; Sun, Q.; Meurs, E.F.; Lin, R.T.; et al. A functional C-terminal TRAF3-binding site in MAVS participates in positive and negative regulation of the IFN antiviral response. Cell Res. 2011, 21, 895–910. [Google Scholar] [CrossRef] [PubMed]
  120. Liu, S.Q.; Chen, J.Q.; Cai, X.; Wu, J.X.; Chen, X.; Wu, Y.T.; Sun, L.J.; Chen, Z.J.J. MAVS recruits multiple ubiquitin E3 ligases to activate antiviral signaling cascades. eLife 2013, 2, e00785. [Google Scholar] [CrossRef]
  121. Fang, R.; Jiang, Q.; Zhou, X.; Wang, C.; Guan, Y.; Tao, J.; Xi, J.; Feng, J.-M.; Jiang, Z. MAVS activates TBK1 and IKKε through TRAFs in NEMO dependent and independent manner. PLoS Pathog. 2017, 13, e1006720. [Google Scholar] [CrossRef] [PubMed]
  122. Sharma, S.; tenOever, B.R.; Grandvaux, N.; Zhou, G.P.; Lin, R.T.; Hiscott, J. Triggering the interferon antiviral response through an IKK-related pathway. Science 2003, 300, 1148–1151. [Google Scholar] [CrossRef]
  123. Fitzgerald, K.A.; McWhirter, S.M.; Faia, K.L.; Rowe, D.C.; Latz, E.; Golenbock, D.T.; Coyle, A.J.; Liao, S.M.; Maniatis, T. IKKε and TBK1 are essential components of the IRF3 signaling pathway. Nat. Immunol. 2003, 4, 491–496. [Google Scholar] [CrossRef]
  124. Schneider, W.M.; Chevillotte, M.D.; Rice, C.M. Interferon-Stimulated Genes: A Complex Web of Host Defenses. Annu. Rev. Immunol. 2014, 32, 513–545. [Google Scholar] [CrossRef]
  125. García, M.A.; Gil, J.; Ventoso, I.; Guerra, S.; Domingo, E.; Rivas, C.; Esteban, M. Impact of protein kinase PKR in cell biology: From antiviral to antiproliferative action. Microbiol. Mol. Biol. Rev. 2006, 70, 1032. [Google Scholar] [CrossRef] [PubMed]
  126. Kim, Y.; Park, J.; Kim, S.; Kim, M.; Kang, M.G.; Kwak, C.; Kang, M.; Kim, B.; Rhee, H.W.; Kim, V.N. PKR Senses Nuclear and Mitochondrial Signals by Interacting with Endogenous Double-Stranded RNAs. Mol. Cell 2018, 71, 1051. [Google Scholar] [CrossRef]
  127. Lemaire, P.A.; Anderson, E.; Lary, J.; Cole, J.L. Mechanism of PKR activation by dsRNA. J. Mol. Biol. 2008, 381, 351–360. [Google Scholar] [CrossRef]
  128. Clemens, M.J.; Elia, A. The double-stranded RNA-dependent protein kinase PKR: Structure and function. J. Interf. Cytok Res. 1997, 17, 503–524. [Google Scholar] [CrossRef]
  129. Gil, J.; Esteban, M. Induction of apoptosis by the dsRNA-dependent protein kinase (PKR): Mechanism of action. Apoptosis 2000, 5, 107–114. [Google Scholar] [CrossRef]
  130. Pajak, A.; Laine, I.; Clemente, P.; El-Fissi, N.; Schober, F.A.; Maffezzini, C.; Calvo-Garrido, J.; Wibom, R.; Filograna, R.; Dhir, A.; et al. Defects of mitochondrial RNA turnover lead to the accumulation of double-stranded RNA. PLoS Genet. 2019, 15, e1008240. [Google Scholar] [CrossRef] [PubMed]
  131. Kim, S.; Tan, S.; Ku, J.; Widowati, T.A.; Ku, D.; Lee, K.; You, K.; Kim, Y. RNA 5-methylcytosine marks mitochondrial double-stranded RNAs for degradation and cytosolic release. Mol. Cell 2024, 84, 2935–2948.e7. [Google Scholar] [CrossRef]
  132. Hooftman, A.; Peace, C.G.; Ryan, D.G.; Day, E.A.; Yang, M.; McGettrick, A.F.; Yin, M.R.; Montano, E.N.; Huo, L.H.; Toller-Kawahisa, J.E.; et al. Macrophage fumarate hydratase restrains mtRNA-mediated interferon production. Nature 2023, 615, 490. [Google Scholar] [CrossRef]
  133. Yoon, J.; Lee, M.; Ali, A.A.; Oh, Y.R.; Choi, Y.S.; Kim, S.; Lee, N.; Jang, S.G.; Park, S.; Chung, J.-H.; et al. Mitochondrial double-stranded RNAs as a pivotal mediator in the pathogenesis of Sjӧgren’s syndrome. Mol. Ther. Nucleic Acids. 2022, 30, 257–269. [Google Scholar] [CrossRef]
  134. López-Polo, V.; Maus, M.; Zacharioudakis, E.; Lafarga, M.; Attolini, C.S.O.; Marques, F.D.M.; Kovatcheva, M.; Gavathiotis, E.; Serrano, M. Release of mitochondrial dsRNA into the cytosol is a key driver of the inflammatory phenotype of senescent cells. Nat. Commun. 2024, 15, 7378. [Google Scholar] [CrossRef]
  135. Victorelli, S.; Salmonowicz, H.; Chapman, J.; Martini, H.; Vizioli, M.G.; Riley, J.S.; Cloix, C.; Hall-Younger, E.; Espindola-Netto, J.M.; Jurk, D.; et al. Apoptotic stress causes mtDNA release during senescence and drives the SASP. Nature 2024, 625, E15. [Google Scholar] [CrossRef]
  136. Nakayama, Y.; Fujiu, K.; Yuki, R.; Oishi, Y.; Morioka, M.S.; Isagawa, T.; Matsuda, J.; Oshima, T.; Matsubara, T.; Sugita, J.; et al. A long noncoding RNA regulates inflammation resolution by mouse macrophages through fatty acid oxidation activation. Proc. Natl. Acad. Sci. USA 2020, 117, 14365–14375. [Google Scholar] [CrossRef]
  137. Zhao, Q.Y.; Liu, J.Y.; Deng, H.; Ma, R.Y.; Liao, J.Y.; Liang, H.X.; Hu, J.X.; Li, J.Q.; Guo, Z.Y.; Cai, J.C.; et al. Targeting Mitochondria-Located circRNA SCAR Alleviates NASH via Reducing mROS Output. Cell 2020, 183, 76. [Google Scholar] [CrossRef]
  138. Liu, X.; Fu, R.; Pan, Y.D.; Meza-Sosa, K.F.; Zhang, Z.B.; Lieberman, J. PNPT1 Release from Mitochondria during Apoptosis Triggers Decay of Poly(A) RNAs. Cell 2018, 174, 187. [Google Scholar] [CrossRef]
  139. Bérouti, M.; Wagner, M.; Greulich, W.; Piseddu, I.; Gärtig, J.; Hansbauer, L.; Müller-Hermes, C.; Heiss, M.; Pichler, A.; Tölke, A.J.; et al. Pseudouridine RNA avoids immune detection through impaired endolysosomal processing and TLR engagement. Cell 2025, 188, 4880–4895.e15. [Google Scholar] [CrossRef]
  140. Ding, Z.C.; Wang, R.; Li, Y.H.; Wang, X.D. MLKL activates the cGAS-STING pathway by releasing mitochondrial DNA upon necroptosis induction. Mol. Cell 2025, 85, 2610–2625.e5. [Google Scholar] [CrossRef]
  141. Newman, L.E.; Shadel, G.S. Mitochondrial DNA Release in Innate Immune Signaling. Annu. Rev. Biochem. 2023, 92, 299–332. [Google Scholar] [CrossRef]
  142. Liao, S.Y.; Chen, L.; Song, Z.Y.; He, H. The fate of damaged mitochondrial DNA in the cell. Biochim. Biophys. Acta-Mol. Cell Res. 2022, 1869, 119233. [Google Scholar] [CrossRef]
  143. Miao, R.; Jiang, C.; Chang, W.Y.; Zhang, H.W.; An, J.S.; Ho, F.; Chen, P.C.; Zhang, H.; Junqueira, C.; Amgalan, D.; et al. Gasdermin D permeabilization of mitochondrial inner and outer membranes accelerates and enhances pyroptosis. Immunity 2023, 56, 2523–2541.e8. [Google Scholar] [CrossRef]
  144. Wang, P.C.; Zhang, L.X.; Chen, S.Y.; Li, R.J.; Liu, P.P.; Li, X.; Luo, H.D.; Huo, Y.J.; Zhang, Z.R.; Cai, Y.Q.; et al. ANT2 functions as a translocon for mitochondrial cross-membrane translocation of RNAs. Cell Res. 2024, 34, 504–521. [Google Scholar] [CrossRef]
  145. Zecchini, V.; Paupe, V.; Herranz-Montoya, I.; Janssen, J.; Wortel, I.M.N.; Morris, J.L.; Ferguson, A.; Chowdury, S.R.; Segarra-Mondejar, M.; Costa, A.S.H.; et al. Fumarate induces vesicular release of mtDNA to drive innate immunity. Nature 2023, 615, 499. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of human mitochondrial DNA (mtDNA) and fundamental stages of mitochondrial RNA (mtRNA) metabolism. The mitochondrial genome encodes 37 genes, including 22 transfer RNAs (tRNAs), 2 ribosomal RNAs (rRNAs), and 13 messenger RNAs (mRNAs). Mitochondrial DNA transcription is initiated by POLRMT with the essential assistance of TFAM and TFB2M. This process generates bidirectional polycistronic transcripts from both the heavy and light strands. These primary transcripts are subsequently processed by RNase P and RNase Z (ELAC2), which cleave at tRNA boundaries to release individual mRNAs, tRNAs, and rRNAs. Following cleavage, the liberated RNAs undergo diverse post-transcriptional modifications catalyzed by specific enzymes. These modifications serve critical functions, including stabilizing RNA structure and facilitating translation. Finally, mitochondrial RNA degradation is mediated by the PNPT1-SUV3 complex and REXO2. The solid line refers to antisense-RNA. The middle dashed line represents the small RNA fragments degraded by PNPT1, and the rightmost dashed line represents the RNA degraded by REXO2. Created in BioRender. Created in BioRender. Yan, C. (2025) https://BioRender.com/a4gafh7.
Figure 1. Schematic representation of human mitochondrial DNA (mtDNA) and fundamental stages of mitochondrial RNA (mtRNA) metabolism. The mitochondrial genome encodes 37 genes, including 22 transfer RNAs (tRNAs), 2 ribosomal RNAs (rRNAs), and 13 messenger RNAs (mRNAs). Mitochondrial DNA transcription is initiated by POLRMT with the essential assistance of TFAM and TFB2M. This process generates bidirectional polycistronic transcripts from both the heavy and light strands. These primary transcripts are subsequently processed by RNase P and RNase Z (ELAC2), which cleave at tRNA boundaries to release individual mRNAs, tRNAs, and rRNAs. Following cleavage, the liberated RNAs undergo diverse post-transcriptional modifications catalyzed by specific enzymes. These modifications serve critical functions, including stabilizing RNA structure and facilitating translation. Finally, mitochondrial RNA degradation is mediated by the PNPT1-SUV3 complex and REXO2. The solid line refers to antisense-RNA. The middle dashed line represents the small RNA fragments degraded by PNPT1, and the rightmost dashed line represents the RNA degraded by REXO2. Created in BioRender. Created in BioRender. Yan, C. (2025) https://BioRender.com/a4gafh7.
Biomolecules 15 01379 g001
Figure 2. Released mtRNA can activate immune responses. Under normal conditions, mt-dsRNA generated within mitochondria is degraded by the PNPT1-SUV3 complex and subsequently processed by REXO2. However, under stress conditions—such as PNPT1 inactivation or NSUN4 deficiency—mt-dsRNA accumulates abnormally. This accumulated mt-dsRNA escapes the mitochondria into the cytosol via channels in the outer mitochondrial membrane, including BAX/BAK, VDAC1, and potentially other unidentified channels or mechanisms like mitochondrial-derived vesicles (MDVs). In the cytosol, the released mt-dsRNA is recognized by sensors including MDA5, RIG-I, PKR, and TLR3. This recognition triggers immune signaling pathways, leading to the production of type I interferons and other inflammatory mediators. All line segments with arrows indicate promotion. The long dashed line represents the dividing line between normal conditions and stress conditions. Created in BioRender. Yan, C. (2025) https://BioRender.com/yb3vr4x.
Figure 2. Released mtRNA can activate immune responses. Under normal conditions, mt-dsRNA generated within mitochondria is degraded by the PNPT1-SUV3 complex and subsequently processed by REXO2. However, under stress conditions—such as PNPT1 inactivation or NSUN4 deficiency—mt-dsRNA accumulates abnormally. This accumulated mt-dsRNA escapes the mitochondria into the cytosol via channels in the outer mitochondrial membrane, including BAX/BAK, VDAC1, and potentially other unidentified channels or mechanisms like mitochondrial-derived vesicles (MDVs). In the cytosol, the released mt-dsRNA is recognized by sensors including MDA5, RIG-I, PKR, and TLR3. This recognition triggers immune signaling pathways, leading to the production of type I interferons and other inflammatory mediators. All line segments with arrows indicate promotion. The long dashed line represents the dividing line between normal conditions and stress conditions. Created in BioRender. Yan, C. (2025) https://BioRender.com/yb3vr4x.
Biomolecules 15 01379 g002
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

Yan, C.; Yu, J.; Lyu, H.; Xiao, S.; Guo, D.; Zhang, Q.; Zhang, R.; Tang, J.; Song, Z.; Zhou, C. Transcription, Maturation and Degradation of Mitochondrial RNA: Implications for Innate Immune Response. Biomolecules 2025, 15, 1379. https://doi.org/10.3390/biom15101379

AMA Style

Yan C, Yu J, Lyu H, Xiao S, Guo D, Zhang Q, Zhang R, Tang J, Song Z, Zhou C. Transcription, Maturation and Degradation of Mitochondrial RNA: Implications for Innate Immune Response. Biomolecules. 2025; 15(10):1379. https://doi.org/10.3390/biom15101379

Chicago/Turabian Style

Yan, Chaojun, Jianglong Yu, Hao Lyu, Shuai Xiao, Dong Guo, Qi Zhang, Rui Zhang, Jingfeng Tang, Zhiyin Song, and Cefan Zhou. 2025. "Transcription, Maturation and Degradation of Mitochondrial RNA: Implications for Innate Immune Response" Biomolecules 15, no. 10: 1379. https://doi.org/10.3390/biom15101379

APA Style

Yan, C., Yu, J., Lyu, H., Xiao, S., Guo, D., Zhang, Q., Zhang, R., Tang, J., Song, Z., & Zhou, C. (2025). Transcription, Maturation and Degradation of Mitochondrial RNA: Implications for Innate Immune Response. Biomolecules, 15(10), 1379. https://doi.org/10.3390/biom15101379

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