Next Article in Journal
Mitochondrial DNA Variants at Low-Level Heteroplasmy and Decreased Copy Numbers in Chronic Kidney Disease (CKD) Tissues with Kidney Cancer
Next Article in Special Issue
From Beach to the Bedside: Harnessing Mitochondrial Function in Human Diseases Using New Marine-Derived Strategies
Previous Article in Journal
Targeting Cathepsin L in Cancer Management: Leveraging Machine Learning, Structure-Based Virtual Screening, and Molecular Dynamics Studies
Previous Article in Special Issue
Mitochondrial Dysfunction in Skeletal Muscle of Rotenone-Induced Rat Model of Parkinson’s Disease: SC-Nanophytosomes as Therapeutic Approach
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Mitochondrial p38 Mitogen-Activated Protein Kinase: Insights into Its Regulation of and Role in LONP1-Deficient Nematodes

by
Eirini Taouktsi
1,2,
Eleni Kyriakou
1,†,
Evangelia Voulgaraki
1,
Dimitris Verganelakis
1,3,
Stefania Krokou
1,
Stamatis Rigas
2,
Gerassimos E. Voutsinas
4 and
Popi Syntichaki
1,*
1
Laboratory of Molecular Genetics of Aging, Biomedical Research Foundation of the Academy of Athens, Center of Basic Research, 11527 Athens, Greece
2
Department of Biotechnology, Agricultural University of Athens, 11855 Athens, Greece
3
Department of Biological Applications & Technology, University of Ioannina, 45500 Ioannina, Greece
4
Laboratory of Molecular Carcinogenesis and Rare Disease Genetics, Institute of Biosciences and Applications, National Center for Scientific Research “Demokritos”, Aghia Paraskevi Attikis, 15341 Athens, Greece
*
Author to whom correspondence should be addressed.
Present Address: Department of Hygiene, Epidemiology and Medical Statistics, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece.
Int. J. Mol. Sci. 2023, 24(24), 17209; https://doi.org/10.3390/ijms242417209
Submission received: 23 October 2023 / Revised: 30 November 2023 / Accepted: 5 December 2023 / Published: 7 December 2023
(This article belongs to the Special Issue Mitochondria in Human Health and Disease 2.0)

Abstract

:
p38 Mitogen-Activated Protein Kinase (MAPK) cascades are central regulators of numerous physiological cellular processes, including stress response signaling. In C. elegans, mitochondrial dysfunction activates a PMK-3/p38 MAPK signaling pathway (MAPKmt), but its functional role still remains elusive. Here, we demonstrate the induction of MAPKmt in worms deficient in the lonp-1 gene, which encodes the worm ortholog of mammalian mitochondrial LonP1. This induction is subjected to negative regulation by the ATFS-1 transcription factor through the CREB-binding protein (CBP) ortholog CBP-3, indicating an interplay between both activated MAPKmt and mitochondrial Unfolded Protein Response (UPRmt) surveillance pathways. Our results also reveal a genetic interaction in lonp-1 mutants between PMK-3 kinase and the ZIP-2 transcription factor. ZIP-2 has an established role in innate immunity but can also modulate the lifespan by maintaining mitochondrial homeostasis during ageing. We show that in lonp-1 animals, ZIP-2 is activated in a PMK-3-dependent manner but does not confer increased survival to pathogenic bacteria. However, deletion of zip-2 or pmk-3 shortens the lifespan of lonp-1 mutants, suggesting a possible crosstalk under conditions of mitochondrial perturbation that influences the ageing process. Furthermore, loss of pmk-3 specifically diminished the extreme heat tolerance of lonp-1 worms, highlighting the crucial role of PMK-3 in the heat shock response upon mitochondrial LONP-1 inactivation.

1. Introduction

The evolutionarily conserved p38 mitogen-activated protein kinase (p38 MAPK) signaling cascade transmits diverse extracellular cues to intracellular targets, regulating mainly stress responses, but also other vital cellular processes, such as cell proliferation, differentiation, immune responses, tumorigenesis and apoptosis [1,2]. The C. elegans genome contains three p38 MAPK homologs, PMK-1, PMK-2 and PMK-3, which are encoded by one polycistronic transcript of an operon under the activity of a broadly expressed promoter [3]. PMK-1 and PMK-2 function redundantly in the nervous system during development [4]. The neuronal functions of PMK-1 and PMK-2 are regulated by a signaling cascade consisting of the adaptor protein TIR-1, which is orthologous to mammalian Sterile α and Armadillo Motif (SARM), and the upstream kinases NSY-1 (MAP3K) and SEK-1 (MAPKK), which are orthologous to mammalian Apoptosis Signal-Regulating Kinase 1 (ASK1) and Mitogen-Activated Protein Kinase Kinase 3/6 (MKK3/6), respectively [5]. In contrast, PMK-3 and its upstream kinases DLK-1 (MAP3K) and MKK-4 or MLK-1(MAPKKs) act in a cascade that regulates synapse formation [6] or axon regrowth after injury [7,8]. PMK-3 also promotes the differentiation of chemosensory BAG neurons, which sense microbe-derived carbon dioxide (CO2) in worms [9,10].
Several studies in C. elegans have uncovered additional roles of PMK-1/p38 MAPK in environmental and cellular stress responses. For example, the PMK-1 pathway has a key role in regulating the expression of innate immune genes in response to various pathogens [11,12]. PMK-1 is the sole effector of the TIR-1/NSY-1/SEK-1 cascade, functioning in the intestines of worms to promote host pathogen resistance [13,14,15,16]. The PMK-1 signaling pathway is also activated in response to abiotic stressors such as cadmium [17,18] and cholesterol deprivation [19]. Additionally, during oxidative stress, it acts in the intestine to phosphorylate SKN-1, the worm ortholog of mammalian Nuclear Factor-Erythroid 2-Related Factor (NRF) transcription factors, mainly regulating transcription of phase II detoxification genes [20,21,22]. Furthermore, PMK-1 and SKN-1 activities have been linked to the heat tolerance of C. elegans, as heat stress induces nuclear translocation of PMK-1 in intestinal cells and depletion of pmk-1 reduces the survival rate of worms at high temperatures [23].
In contrast to PMK-1, there is little knowledge about the role of the PMK-3 signaling pathway in stress responses. A mitochondrial MAPK (MAPKmt) cascade defined by DLK-1 (MAP3K), SEK-3 (MAPKK) and PMK-3 kinases was identified to be required for lifespan extension of C. elegans mutants with disrupted mitochondrial activity [24]. It seems that the PMK-3 pathway defines another cellular retrograde response that is activated in complement to the mitochondrial unfolded protein response (UPRmt), a conserved surveillance mechanism by which metazoans respond to mitochondrial stress [25]. During UPRmt activation, an increased unfolded/misfolded protein load inside the mitochondria redirects the basic leucine zipper (bZIP) transcription factor ATFS-1 from the matrix to the nucleus, regulating the expression of both mitochondrial protective genes and metabolism-related genes [25,26]. In the absence of mitochondrial stress, ATFS-1 is trafficked to the mitochondrial matrix and rapidly degraded by the ATP-dependent protease LONP-1, the ortholog of mammalian LonP1 [25].
LonP1 protease belongs to the ATPase associated with diverse cellular activities (AAA+) family with major roles in mitochondrial protein quality control [27,28,29]. We have previously shown that deletion of the lonp-1 gene in C. elegans increases endogenous levels of ROS, impacts the mitochondrial function and impairs physiological processes, such as growth, behavior and lifespan [30]. Loss of LONP-1 induces the UPRmt, while the accompanied activation of several cytoplasmic UPR (UPRcyt) responses was shown to increase the resistance of mutants to heat and other forms of stress. In addition to worms, genetic or pharmacological inhibition of human LonP1 in cancer cells induces similar mitochondrial and cytoplasmic UPR mechanisms [30]. Although the induction of such UPR pathways could result in lifespan extension of C. elegans, the short-lived phenotype of the lonp-1 mutants implies the existence of additional factors/mechanisms that modulate their lifespan and possibly their viability under adverse environmental conditions. In this study, we demonstrated the induction of the MAPKmt signaling pathway in lonp-1 mutants and we examined its relationship with other mitochondrial surveillance mechanisms, in addition to the effects of its disruption on physiological functions of lonp-1-deficient animals. The described relationships and effects might be evolutionarily conserved, thus shedding some light on human health and mitochondrial dysfunction-related diseases.

2. Results

2.1. The MAPKmt Pathway Is Active in Lonp-1-Deficient Mutants

Mitochondrial dysfunction caused by disruption of the mitochondrial electron transport chain, such as mutations in the Rieske iron-sulfur polypeptide 1 (isp-1) of complex III, has previously been shown to activate the MAPKmt pathway, which consists of DLK-1, SEK-3, and PMK-3 kinases and induces transcription of the tbb-6 gene [24] (Figure 1A).
tbb-6 encodes for a structurally diverged member of the beta-tubulin family in C. elegans [31], with no reported specific functions or overt mutant phenotypes. We examined the expression levels of tbb-6 in synchronized young adults of wild-type (wt) and lonp-1 knockout strains and we observed elevated mRNA levels of the endogenous tbb-6 gene (Figure 1B), as well as a strong induction of a transcriptional reporter tbb-6p::gfp (Figure 1C) in the intestine and pharyngeal muscles of lonp-1 compared to wt animals. Mild exposure to isp-1(RNAi) further increased the fluorescence of the reporter in lonp-1, indicating a strong additive mitochondrial dysfunction in lonp-1;isp-1(RNAi) adults (interaction p < 0.0005 in a two-way ANOVA; Figure S1A). In addition to tbb-6, the C. elegans beta-tubulin family comprises five more members, of which tbb-1 and tbb-2 are ubiquitously expressed and partially redundant, ben-1/tbb-5 is broadly expressed in the nervous system, and mec-7/tbb-3 and tbb-4 are expressed only in touch receptor neurons [31]. The expressions of all three major beta-tubulins, tbb-1, tbb-2, and ben-1, were not changed in lonp-1 mutant animals (Figure S1B), confirming a specific, yet unidentified, role of TBB-6 in mitochondrial dysfunction. However, the expression levels of the tbb-6 gene and reporter were found to severely drop upon RNAi-mediated disruption of DLK-1 and SEK-3 or deletion of PMK-3 in both wt and lonp-1 mutants (Figure 1D,E and Figure S2). Thus, we concluded that MAPKmt signaling regulates both the basal and the lonp-1-induced expression of the tbb-6 gene.

2.2. ATFS-1 Counteracts Activation of the MAPKmt Pathway upon Lonp-1 Inactivation

Mitochondrial perturbation by LONP-1 deficiency has been demonstrated to elicit activation of several stress-responsive transcription factors, including SKN-1 and DAF-16, the worm orthologs of the mammalian NRF and Forkhead Box O (FOXO) factors, respectively [30]. Since the promoter of tbb-6 contains putative binding sites for SKN-1 and DAF-16 transcription factors (WormBase version WS289), we tested their requirement for tbb-6 induction in lonp-1 mutants. RNAi targeting SKN-1 or DAF-16 did not reduce fluorescence of the tbb-6p::gfp reporter or tbb-6 mRNA levels (Figure S3), suggesting that these factors are not involved in the expression of tbb-6 in lonp-1 adults. Interestingly, genetic loss of atfs-1 by the gk3094 allele led to high tbb-6 mRNA levels in an otherwise wt background (Figure 2A), and RNAi-induced depletion of atfs-1 in a lonp-1 background further increased the expression of the tbb-6 gene or the tbb-6p::gfp reporter (Figure 2A,B).
These results support the notion that ATFS-1 counteracts the activity of the MAPKmt pathway, as has been previously suggested for isp-1(qm150) mutants [24]. However, when two other UPRmt-associated factors, namely DVE-1 and UBL-5 [32], were knocked down by RNAi, no difference in the signal of the tbb-6 reporter was detected (Figure S4), suggesting that ATFS-1 exerts a repressive function independently of the canonical UPRmt signaling branch. The repressive function of ATFS-1 on tbb-6 expression occurs upstream of PMK-3, as atfs-1(RNAi) was not able to increase the tbb-6p::gfp fluorescence in lonp-1;pmk-3 double mutants (Figure 2B).

2.3. ATFS-1 Suppresses the Induction of cbp-3 in Lonp-1 Mutants

It has been reported that tbb-6 induction in isp-1 mutant worms depends on cbp-3/F40F12.7, a homolog of the mammalian acetyltransferase CBP/p300 [24]. However, cbp-3 is probably a pseudogene of cbp-1, bearing only the kinase-inducible domain interacting domain (KIX) and missing the histone acetyltransferase (HAT) domain of CBP-1 [33]. Thus, in contrast to cbp-1(RNAi), we showed that RNAi against cbp-3 does not cause developmental defects and arrest in worms, possibly due to lack of the HAT domain. When lonp-1 mutant adults were subjected to cbp-3(RNAi), the expression levels of tbb-6p::gfp or the endogenous tbb-6 gene were significantly decreased (Figure 3A,B), supporting the role of cbp-3 in the induction of tbb-6 in a lonp-1 background.
We further showed that the mRNA levels of cbp-3, and not those of cbp-1, were up-regulated in lonp-1 mutants, independently of PMK-3 function (Figure 3C). Although the expression of cbp-3 has been previously shown to increase upon induction of UPRmt in an ATFS-1-dependent manner [25], we found that depletion of atfs-1 enhances, rather than decreases, the expression of cbp-3 in lonp-1 mutants (Figure 3D), similarly to tbb-6. Depletion of SKN-1 had no effect on cbp-3 transcript levels (Figure 3D), which is consistent with the lack of effects of skn-1(RNAi) in tbb-6 mRNA levels. Thus, our data suggest that ATFS-1 suppresses the induction of cbp-3 in lonp-1 mutants and imply a role of the KIX domain of CBP-3 in the activation of the MAPKmt pathway, placing it upstream of PMK-3/TBB-6 signaling.

2.4. Induction of the MAPKmt Pathway in Lonp-1 Mutants Is Not MAD Pathway-Dependent

Mitochondrial dysfunction in yeast strongly activates the mitochondria-associated degradation (MAD) pathway [34], and inhibition of MAD components could block tbb-6p::gfp induction in isp-1 mutants [24]. The MAD pathway mediates ubiquitination and degradation of misfolded or damaged mitochondrial proteins under mitochondrial stress, analogously to the endoplasmic reticulum (ER)-associated degradation pathway (ERAD). Physical interaction of CBP-3 with proteins involved in protein ubiquitination, such as F57C12.2 [35,36] and ubq-1 [37], has been reported; thus, we wondered whether CBP-3 functions with the MAD pathway to enable induction of MAPKmt in lonp-1 mutants. C. elegans has two orthologs of a highly conserved AAA+ ATPase, named p97/VCP/CDC48 in mammals, which direct ubiquitinated proteins from cellular membranes to the 26S proteasome for degradation [38,39,40]. The worm orthologs, CDC-48.1 and CDC-48.2, share 88% identity over the entire p97 protein, and both have essential and redundant functions [41]. We introduced a null mutation of cdc-48.2 into lonp-1 worms, but did not find a significant change in fluorescence of the reporter tbb-6p::gfp or the mRNA levels of tbb-6 gene (Figure 4A). In a similar manner, depletion of cdc-48.1 through RNAi, alone or in combination with cdc-48.2(tm659), did not reduce the expression of tbb-6 in lonp-1 mutants (Figure 4B). These results indicate that the activity of the MAD pathway is not a prerequisite for MAPKmt induction, at least in lonp-1-deficient worms, and cbp-3 possibly influences PMK-3 activity through mechanisms independent of MAD signaling.

2.5. Loss of Lonp-1 Induces the ZIP-2 Innate Immune Response Effector

Several studies of C. elegans have established that disruption of essential core processes, including mitochondrial function, can induce innate immune responses in worms [42,43]. In particular, UPRmt has been shown to induce a subset of ATFS-1-dependent innate immunity genes, providing resistance to pathogenic bacteria Pseudomonas aeruginosa, independently of PMK-1/p38 MAPK [44]. One such gene is the infection response gene-1 (irg-1), a target of the bZIP transcription factor ZIP-2 [42,45,46], which was reported to be upregulated by ATFS-1 upon UPRmt [25,44]. The expression of irg-1 and zip-2 was examined and was found to be significantly induced in lonp-1 mutants compared to wt animals (Figure 5A). In addition to irg-1, the infection response gene-2 (irg-2), another ZIP-2 target [42,45], was upregulated upon lonp-1 deletion (Figure 5A). Using a zip-2(ok3730) loss-of-function allele, we verified that ZIP-2 was required for basal expression of both irg-1 and irg-2, whereas in a lonp-1 mutant background, ZIP-2 was necessary only for irg-2 induction (Figure 5A). Instead, irg-1 could be upregulated in lonp-1 worms by ZIP-2-independent mechanisms, as was previously reported for other conditions, e.g., suppression of translation [42].
Nevertheless, mitochondrial damage caused during infection by P. aeruginosa elicits a host defense, mediated in part by the ZIP-2 transcription factor, to promote organismal survival [47,48]. However, the increased activity of ATFS-1 and ZIP-2 in the lonp-1 background was not sufficient to promote host resistance to infection by the P. aeruginosa strain PA14, and lonp-1 adults were significantly susceptible to the pathogen compared to wt animals (Figure 5B). Given that ATFS-1 activity counteracts the activation of the MAPKmt pathway, we tested whether genetic disruption of pmk-3 could enhance the host defense response to PA14. As shown in Figure 5B, loss of pmk-3 did not alter the hypersensitive phenotype of lonp-1 mutants to pathogenic bacteria, although it had a negative impact on the immune response of wt worms. In spite of that, ZIP-2-dependent expression of irg-1 and irg-2 genes was compromised in both pmk-3 and lonp-1;pmk-3 mutants, similarly to zip-2 deletion (Figure 5A). These results suggest a genetic interaction between PMK-3 and ZIP-2 when lonp-1 is missing, which determines the activity of ZIP-2 and affects other functions than the innate immune response.

2.6. Disruption of the MAPKmt Pathway Influences Lonp-1 Mutants’ Lifespan and Stress Responses

Induction of the ZIP-2 pathway provides surveillance for several core cellular processes, and can also delay the ageing process by preserving mitochondrial homeostasis in aged worms [49]. Moreover, the extended lifespan of some, but not all, long-lived mitochondrial mutants was shown to require an active MAPKmt pathway [24]. Considering the above genetic interaction between ZIP-2 and PMK-3, we examined the effects of disruption of each pathway on the lifespan of lonp-1 mutants. Although worms deficient in the lonp-1 gene display a shorter lifespan than the wt [30], deletion of either zip-2 or pmk-3 severely reduced the lifespan of lonp-1 mutants, without affecting the normal lifespan of wt worms (Figure 6A and Table S3). This shortening of lifespans was not observed when pmk-1/p38 MAPK was deleted in the lonp-1 mutant background, with loss of pmk-1 to reduce only the brood size of both wt and lonp-1 strains (Figure S5A,B and Table S3). Furthermore, deletion of pmk-3, pmk-1 and zip-2 (Figure S5C) did not have any impact on the growth of wt or lonp-1 animals.
Worms bearing deficiency in the lonp-1 gene are short-lived relative to the wt, but exhibit an increased resilience in some forms of exogenous stress, for instance, to specific oxidants or heat shock [30]. Here, we showed that, similarly to pmk-1 deletion, loss of pmk-3 renders both wt and lonp-1 worms sensitive to oxidative stress induced by the organic peroxide tert-butyl hydroperoxide (tBHP) (Figure 6B and Figure S6A). Treatment of worms with antimycin, an inhibitor of complex III of the electron transport chain, revealed a general requirement for pmk-3 to confront superoxide anion generation in mitochondria (Figure 6B), while pmk-1 was required only in lonp-1-deficient animals (Figure S6A). These results suggest that PMK-3 is important for both wt and lonp-1 adults to cope with oxidative stress. Moreover, deletion of pmk-3 severely reduced the high survival rate of lonp-1 mutants at 35 °C (Figure 6B), while loss of pmk-1 had no effect on their extreme heat tolerance, with the double lonp-1;pmk-1 mutants being similarly tolerant to single lonp-1 mutants (Figure S6A). However, in a wt background, pmk-1 deletion had a greater negative impact on thermotolerance compared to pmk-3 deletion. Further studies are needed to address the key role of PMK-3 in the heat resistance of lonp-1, but it does not seem to involve tbb-6 or zip-2 functions (Figure S6B,C). Overall, our data support a central role of PMK-3/p38 MAPK in mediating the lifespan length and stress resistance under conditions of lonp-1-induced mitochondrial perturbations.

3. Discussion

In humans, mutations of the LonP1 gene are associated with a broad spectrum of diseases, including a rare multisystem developmental disorder known as CODAS (Cerebral, Ocular, Dental, Auricular, and Skeletal anomalies) syndrome, as well as neurodegeneration and cancer [27,28,29,50]. The mammalian protease LonP1 is a multifunctional enzyme with essential roles during development of cardiomyocyte and skeletal muscle cells [51,52]. Also, acting as both a chaperone and a protease, LonP1 plays a crucial role in protein quality control, preserving mitochondrial homeostasis in adults. An age-related decline in the expression and activity of LonP1 seems to contribute to structural and functional impairment of organs with age [29,51]. While genetic disruption of the LonP1 gene in mice and Drosophila results in embryonic lethality [53,54,55], C. elegans lonp-1 knockout mutants are viable but exhibit disturbed mitochondrial and ROS homeostasis, resulting in impaired growth and a shortened lifespan [30]. lonp-1-deficient worms are extremely resistant to specific oxidants and heat stress due to induction of both mitochondrial and cytosolic stress signaling pathways. These include UPRmt, where increased activity of the ATFS-1 transcription factor was found to be essential for normal development of lonp-1 mutants and determines the resistance of lonp-1 adults to external oxidative stress [30]. Interestingly, lack of LONP-1 was recently reported to induce constitutive formation of mitochondrial stress granules, perhaps as an adaptive response [56]. Moreover, ATFS-1 activity in long-lived isp-1 mitochondrial mutants could prevent the induction of another mitochondrial stress signaling pathway named MAPKmt, in which PMK-3/p38 MAPK upregulates the expression of the tbb-6 beta-tubulin target gene in the intestines [24]. MAPKs are potent regulators of microtubule dynamics in mammals, and the DLK-1/PMK-3 pathway in C. elegans can regulate microtubule dynamics and endocytosis in neurons [57,58]. Although the exact role of TBB-6 in intestinal cells is still unknown, it has been suggested that it may function to enhance ADP entry into mitochondria during mitochondrial stress [24].
Here, we demonstrated the induction of the MAPKmt pathway in lonp-1 mutants, which is consistent with the presence of a disturbed mitochondrial network and an accumulation of ROS in these animals. Based on our results discussed below, a schematic representation of the genetic interactions and molecular functions of MAPKmt in worms deficient in the lonp-1 gene is shown in Figure 7. Depletion of atfs-1 further enhances the expression of the tbb-6 marker, supporting the notion that ATFS-1 suppresses MAPKmt activity, probably to fine-tune it. One regulator of ATFS-1 in PMK-3 activity may be the CBP ortholog CBP-3, which contains only the kinase-inducible domain interacting domain [33], and is required for tbb-6 induction in isp-1 mutants [24]. We found that cbp-3 is upregulated in lonp-1 worms and ATFS-1 suppresses this induction upstream of PMK-3/TTB-6 signaling. The combination of such protein interaction and phosphorylation events could be important for orchestrating the cellular processes and changing the metabolic status under two opposing signals in response to mitochondrial stress. For example, under conditions of moderate mitochondrial dysfunction, C. elegans can adapt and often exhibits an altered metabolism and an increased lifespan [59]. However, the ageing process can be accelerated when the activity of the different mitochondrial protein quality control systems is not in balance. In support of this, activity of the MAPKmt pathway is required for the lifespan extension of some mitochondrial mutants [24]. We have shown that disruption of atfs-1 in lonp-1 adults reduced their lifespan [30] and here, deletion of pmk-3 also shortened the lifespan of lonp-1 animals. Moreover, our data as presented below uncovered a genetic interaction between PMK-3 and ZIP-2, which could influence the lifespan of lonp-1 mutants.
ZIP-2 is a bZIP transcription factor with an established role in innate immunity [42,45,46]. Although mitochondrial dysfunction can induce immune responses in worms, mediated in part by ATFS-1 and ZIP-2 [44,47,48], the observed activation of ZIP-2 in lonp-1 mutants was not sufficient to increase survival of mutants against pathogenic bacteria PA14. Moreover, the ZIP-2-dependent transcription of irg-1 and irg-2 target genes was abolished after depletion of pmk-3, signifying a requirement of the PMK-3 kinase for both basal and induced activity of the ZIP-2 factor, even though PMK-3 is not involved in innate immunity. Separate from its role in immunity, ZIP-2 is activated in response to age-associated mitochondrial dysfunction, independently of ATFS-1, and delays aging phenotypes by preserving mitochondrial homeostasis in aged worms [49]. In addition to this, ZIP-2 is activated and mediates lifespan extension under various dietary restriction conditions [60,61]. Since deletion of either zip-2 or pmk-3 significantly reduced the lifespan of lonp-1 mutants, we speculate that this novel crosstalk between MAPKmt and ZIP-2 is important in modulating the ageing process under mitochondrial dysfunction. This is strengthened by the observation that the DLK-1/PMK-3 signaling pathway declines with age, and this is overcome by mild mitochondrial stress [62]. Future studies investigating the underlying molecular mechanism and transcriptional targets are expected to shed light on the relevant ageing modulators.
We have previously shown that knockout of lonp-1 induces a subset of stress-responsive genes associated with protective mechanisms, thereby safeguarding the mitochondrial and cytosolic proteome to defend the mitochondrial function [30]. Thus, here, we evaluated the consequences of pmk-3 deletion on the resistance of lonp-1 mutant adults to exogenous stresses. We confirmed that the activity of PMK-3 is indispensable for both oxidative and heat stress responses. More specifically, loss of PMK-3 significantly diminished the extreme heat resistance of lonp-1 mutants, while deletion of pmk-1 did not block this response. However, thermotolerance of wt animals was reduced when pmk-1 was missing, which is consistent with a previous work, where heat was found to induce nuclear translocation of PMK-1 [23]. It has been suggested that PMK-1 might contribute to the elevated expression of hsp and other chaperone genes through hyperphosphorylation of heat shock factor I (HSF-1), analogously to the stress activation of mammalian HSF1 [63]. HSF-1 is the single worm homolog of the four mammalian HSFs, and its functions in thermal stress response are tightly regulated by post-translational modifications [64]. Given that the extreme thermotolerance of lonp-1 mutants requires HSF-1 activity [30] and PMK-3, but not PMK-1 (this study), we propose that PMK-3 kinase mediates the function of HSF-1 or its regulators in lonp-1 mutants in response to heat stress. These relationships might be evolutionarily conserved, as we have shown that genetic or pharmacological inhibition of human LonP1 in cancer cells induces the expression of genes involved in the heat shock response (HSR) [30]. Interestingly, new evidence exists for nuclear localization of mitochondrial LonP1 in mammalian cells, and this translocation is increased under heat stress, up-regulating HSF1 target genes [65]. Thus, it was suggested that LonP1 modulates the heat shock response through interaction with and possible degradation of HSF1 in the nucleus [65]. It is tempting to speculate that mammalian p38 MAPK could have a similar regulatory role with PMK-3 on LonP1 and HSF1 functions. Although in mammals there are four p38 isoforms that are highly similar to PMK-1 in worms, they exhibit differential expression/activation patterns and specific downstream effectors [1,66], raising the possibility that they may fulfill analogous cellular functions to the PMK-3 kinase in worms, promoting organismal fitness and stress responses.

4. Materials and Methods

4.1. C. elegans Strains and Culture Conditions

C. elegans strains were maintained at 20 °C on nematode growth medium (NGM) plates seeded with Escherichia coli OP50 as a food source. Whenever it was necessary, NGM plates with UV-killed bacteria supplemented with 40 μg/mL 5-fluoro-2′-deoxyuridine (FUdR; Sigma-Aldrich, St. Louis, MO, USA) were used to prevent progeny growth. All strains used in this study are listed in Table S1.

4.2. RNA Interference

RNAi experiments were conducted at 20 °C on NGM plates seeded with E. coli HT115 (DE3) bacteria, transformed with the indicated RNAi construct (primers listed in Table S2) or the appropriate empty vector (plasmid L4440 or T444T, Addgene, Watertown, MA, USA). Plates containing 2 mM isopropyl b-D-1-thiogalactopyranoside (IPTG; Applichem GmbH, Darmstadt, Germany) were incubated at 37 °C overnight to induce RNAi. Worms were synchronized from eggs on RNAi plates at 20 °C and transferred onto new RNAi plates when they reached L4 or the young prefertile adult stage (YA) for analysis. For post-developmental RNAi experiments, L4 to YA worms raised on OP50 plates were transferred onto RNAi plates for the indicated time.

4.3. Microscopic Analysis

Age-synchronized transgenic worms expressing the indicated fluorescence stress reporter were grown on OP50 or RNAi plates at 20 °C, and a microscopic analysis was performed in 1-day-old adults, considering the developmental delay in lonp-1 mutants. Worms were immobilized with levamisole (Sigma-Aldrich, St Louis, MO, USA) and mounted on 2% agarose pads on glass microscope slides. Images of worms were captured via fluorescent microscopy using a Leica DMRA upright fluorescent microscope equipped with a Hamamatsu ORCA-flash 4.0 camera with a 10× or 40× objective lens. All strains were assayed in parallel and microscopy settings were kept the same in each experiment. Fluorescence was measured as the average pixel intensity with ImageJ 1.52p (Fiji) [67] in the whole captured worm image or in standardized regions of the intestine. At least 50 worms per strain and condition were measured in three biological replicates, and the mean of calculated values was plotted as the mean fluorescence intensity ± SEM. Representative fluorescence and bright field micrographs are shown for each genotype and condition.

4.4. Growth Rate and Fecundity Assays

Worms were synchronized by placing 10 gravid adults of each strain on NGM plates with adequate food (OP50) and they were allowed to lay eggs for 2–3 h before removing. Eggs were left to hatch at 20 °C, and the number of progenies in each developmental stage after 68 h was scored. The total brood size was determined at 20 °C by picking 10 L4 stage nematodes from an NGM plate and placing one nematode onto separate individual NGM plates with OP50. Individual animals were transferred onto new plates daily until the end of the spawning period of worms. The total number of F1 progeny hatched in each plate for each individual was counted.

4.5. Lifespan Analysis

Animals were synchronized by egg-laying, and 100–150 healthy L4 larvae of each strain, were transferred to three NGM plates (approximately 40 per plate) seeded with OP50 bacteria, at 20 °C. Animals were moved to fresh plates every 2–4 days and survival was scored daily by gentle prodding. Worms that failed to respond were considered dead, whereas worms raptured, burrowed into the agar, bagged (that displayed internal hatching of eggs), or animals that crawled off the plates were censored in the analysis. Statistical analysis was performed by comparing each population to the appropriate control and p- values were determined using the log-rank (Mantel-Cox) test. Replicates were carried out as indicated in Table S3.

4.6. Stress Sensitivity Assays

In all assays, animals were synchronized by egg laying on NGM plates with OP50 bacteria at 20 °C. Heat shock assays were performed by shifting 1-day-old adults onto three fresh plates containing approximately 30 animals per plate from 20 °C to 35 °C for the indicated time points. The percentage of alive animals was scored following 16 h of recovery at 20 °C by assessing the response to gentle touch. For the tBHP-induced oxidative stress assay, 1-day-old adult worms were placed on freshly prepared tBHP plates [30] containing 10 mM tBHP (from a stock solution of 70% in water, Sigma-Aldrich, St Louis, MO, USA) devoid of bacteria. During the initial two hours, worms were periodically relocated to the center of the plate, as they tried to “escape” from the plate and avoid tBHP, and survival was counted every hour. For antimycin A (Sigma-Aldrich, St Louis, MO, USA)-induced oxidative stress, synchronized L4 larvae of each strain were placed on plates containing 40 μM FUdR for 24 h. The next day, approximately 30 worms per plate were transferred onto fresh plates without FUdR seeded with UV-killed bacteria containing 40 μM antimycin A. Survival was monitored after 24 h on three plates per strain. At least three independent experiments were performed for each stress assay. Resilience against the human opportunistic pathogen P. aeruginosa was tested using the pathogen infection assay, performed by SunyBiotech (https://www.sunybiotech.com, accessed on 1 February 2023). In brief, age-synchronized worms of the indicated genotype were grown at 20 °C on NGM plates seeded with OP50 bacteria. Approximately 80 of the synchronized young adults of each strain were transferred to fresh FUdR-containing plates seeded with the P. aeruginosa strain PA14. Viability was scored daily and p-values were determined using the log-rank (Mantel-Cox) test.

4.7. RNA Extraction and Quantitative Reverse Transcription PCR (qRT-PCR)

Total RNA was extracted from frozen worm pellets, each containing 200–300 worms, as described previously [30]. In each experimental set, a minimum of three biological replicate samples were collected and analyzed independently. Relative mRNA quantities were determined using the comparative Ct method for quantification, with each sample independently normalized to the endogenous reference gene (ama-1). Gene expression data are presented as the average fold change of all biological replicates relative to the control. The primer sequences used for qRT-PCR are shown in Table S2.

4.8. Statistics

Graphs were constructed and statistical analyses were performed in GraphPad Prism version 8.0.0 for Windows (GraphPad Software, San Diego, CA, USA, www.graphpad.com/scientificsoftware/prism, accessed on 10 December 2021). Statistical analyses were performed by comparing each sample to the appropriate control in the same condition and p-values were determined by a Student’s t-test and depicted as follows: **** p < 0.0001; *** p = 0.0001–0.001; ** p = 0.001–0.01; * p = 0.01–0.05; ns indicates not significant with a p-value ≥ 0.05. For analyses involving multiple strains and conditions, a two-way ANOVA with a Tukey’s multiple comparisons analysis was used to assess the significance and differences between groups.

5. Conclusions

In C. elegans, the lonp-1 gene encodes a mitochondrial AAA+ protease, which has been shown to play a crucial role in protein quality control across species. In this work, we found that worms deficient in lonp-1 have an activated mitochondrial PMK-3/p38 MAPK signaling pathway. In lonp-1 mutants, the enhanced activity of the ATFS-1 transcription factor counteracted activation of the MAPKmt pathway, pointing to the existence of interconnected and fine-tuned defense mechanisms in response to mitochondrial stress. In support of this notion, both ATFS-1 and PMK-3 activities positively regulated the lonp-1 mutants’ lifespan and oxidative stress response. In addition to this, we demonstrated that PMK-3 was required for both the basic and lonp-1-induced transcriptional activity of ZIP-2, a bZIP transcription factor that has previously been shown to preserve mitochondrial homeostasis in aged worms independently of innate immunity. Since loss of ZIP-2 activity reduced the lifespan of lonp-1 animals, we suggest that this genetic interaction between PMK-3 and ZIP-2 can influence the ageing process in the face of mitochondrial damage. Finally, the extreme thermotolerance of lonp-1 mutants was found to require PMK-3 activity, uncovering a new role of MAPKmt in the heat stress response when LONP-1 is missing.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms242417209/s1.

Author Contributions

Conceptualization, P.S.; formal analysis, E.T.; funding acquisition, P.S. and S.R.; investigation, E.T., E.K., E.V., D.V. and S.K.; methodology, E.T. and E.K.; project administration, P.S.; resources, S.R., G.E.V. and P.S.; supervision, S.R., G.E.V. and P.S.; visualization, E.T. and E.K.; writing—original draft, P.S.; writing—review and editing, E.T., E.K., S.R., G.E.V. and P.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research work was supported by the Hellenic Foundation for Research and Innovation (H.F.R.I.) under the “First Call for H.F.R.I. Research Projects to support Faculty members and Researchers and the procurement of high-cost research equipment grant” (Project Number: 1611). S.R. also acknowledges Fondation Santé for funding (Biomedical Research Grant 2023).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article and Supplementary Materials.

Acknowledgments

Some strains were provided by the Caenorhabditis Genetics Center (CGC, University of Minnesota, USA), which is supported by NIH Office of Research Infrastructure Programs (P40 OD010440). We are grateful to all the members of the PS and GV laboratories for their contribution and critical input. We would like to thank the BRFAA Imaging Unit for access to and technical assistance with microscopy.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

MAPKMitogen-Activated Protein Kinase
MAPKKMitogen-Activated Protein Kinase Kinase
MAP3KMitogen-Activated Protein Kinase Kinase Kinase
MAPKmtMitochondrial MAPK
UPRmtMitochondrial Unfolded Protein Response
UPRcytCytoplasmic Unfolded Protein Response
ATFActivating Transcription Factor
SARMSterile α and Armadillo Motif
ASK1Apoptosis Signal-Regulating Kinase 1
MKKMitogen-Activated Protein Kinase Kinase
NRFNuclear Factor-Erythroid 2-Related Factor
FOXOForkhead Box O
CBPCREB-Binding Protein
MADMitochondrial-Associated Degradation
bZIP Basic Leucine Zipper
RNAiRNA Interference
tBHPTert-Butyl Hydroperoxide
hspHeat Shock Protein
HSFHeat Shock Factor
HSRHeat Shock Response

References

  1. Maik-Rachline, G.; Lifshits, L.; Seger, R. Nuclear P38: Roles in Physiological and Pathological Processes and Regulation of Nuclear Translocation. Int. J. Mol. Sci. 2020, 21, 6102. [Google Scholar] [CrossRef] [PubMed]
  2. Plotnikov, A.; Zehorai, E.; Procaccia, S.; Seger, R. The MAPK cascades: Signaling components, nuclear roles and mechanisms of nuclear translocation. Biochim. Biophys. Acta (BBA)—Mol. Cell Res. 2011, 1813, 1619–1633. [Google Scholar] [CrossRef] [PubMed]
  3. Andrusiak, M.G.; Jin, Y. Context Specificity of Stress-activated Mitogen-activated Protein (MAP) Kinase Signaling: The Story as Told by Caenorhabditis elegans. J. Biol. Chem. 2016, 291, 7796–7804. [Google Scholar] [CrossRef] [PubMed]
  4. Pagano, D.J.; Kingston, E.R.; Kim, D.H. Tissue expression pattern of PMK-2 p38 MAPK is established by the miR-58 family in C. elegans. PLoS Genet. 2015, 11, e1004997. [Google Scholar] [CrossRef] [PubMed]
  5. Chuang, C.F.; Bargmann, C.I. A Toll-interleukin 1 repeat protein at the synapse specifies asymmetric odorant receptor expression via ASK1 MAPKKK signaling. Genes Dev. 2005, 19, 270–281. [Google Scholar] [CrossRef] [PubMed]
  6. Nakata, K.; Abrams, B.; Grill, B.; Goncharov, A.; Huang, X.; Chisholm, A.D.; Jin, Y. Regulation of a DLK-1 and p38 MAP kinase pathway by the ubiquitin ligase RPM-1 is required for presynaptic development. Cell 2005, 120, 407–420. [Google Scholar] [CrossRef] [PubMed]
  7. Hammarlund, M.; Nix, P.; Hauth, L.; Jorgensen, E.M.; Bastiani, M. Axon regeneration requires a conserved MAP kinase pathway. Science 2009, 323, 802–806. [Google Scholar] [CrossRef] [PubMed]
  8. Yan, D.; Wu, Z.; Chisholm, A.D.; Jin, Y. The DLK-1 kinase promotes mRNA stability and local translation in C. elegans synapses and axon regeneration. Cell 2009, 138, 1005–1018. [Google Scholar] [CrossRef]
  9. Brandt, J.P.; Ringstad, N. Toll-like Receptor Signaling Promotes Development and Function of Sensory Neurons Required for a C. elegans Pathogen-Avoidance Behavior. Curr. Biol. 2015, 25, 2228–2237. [Google Scholar] [CrossRef]
  10. Horowitz, L.B.; Brandt, J.P.; Ringstad, N. Repression of an activity-dependent autocrine insulin signal is required for sensory neuron development in C. elegans. Development 2019, 146, dev182873. [Google Scholar] [CrossRef]
  11. Kim, D.H.; Feinbaum, R.; Alloing, G.; Emerson, F.E.; Garsin, D.A.; Inoue, H.; Tanaka-Hino, M.; Hisamoto, N.; Matsumoto, K.; Tan, M.-W.; et al. A Conserved p38 MAP Kinase Pathway in Caenorhabditis elegans Innate Immunity. Science 2002, 297, 623–626. [Google Scholar] [CrossRef] [PubMed]
  12. Troemel, E.R.; Chu, S.W.; Reinke, V.; Lee, S.S.; Ausubel, F.M.; Kim, D.H. p38 MAPK regulates expression of immune response genes and contributes to longevity in C. elegans. PLoS Genet. 2006, 2, e183. [Google Scholar] [CrossRef] [PubMed]
  13. Couillault, C.; Pujol, N.; Reboul, J.; Sabatier, L.; Guichou, J.F.; Kohara, Y.; Ewbank, J.J. TLR-independent control of innate immunity in Caenorhabditis elegans by the TIR domain adaptor protein TIR-1, an ortholog of human SARM. Nat. Immunol. 2004, 5, 488–494. [Google Scholar] [CrossRef] [PubMed]
  14. Liberati, N.T.; Fitzgerald, K.A.; Kim, D.H.; Feinbaum, R.; Golenbock, D.T.; Ausubel, F.M. Requirement for a conserved Toll/interleukin-1 resistance domain protein in the Caenorhabditis elegans immune response. Proc. Natl. Acad. Sci. USA 2004, 101, 6593–6598. [Google Scholar] [CrossRef] [PubMed]
  15. Pukkila-Worley, R.; Ausubel, F.M. Immune defense mechanisms in the Caenorhabditis elegans intestinal epithelium. Curr. Opin. Immunol. 2012, 24, 3–9. [Google Scholar] [CrossRef] [PubMed]
  16. Shivers, R.P.; Kooistra, T.; Chu, S.W.; Pagano, D.J.; Kim, D.H. Tissue-specific activities of an immune signaling module regulate physiological responses to pathogenic and nutritional bacteria in C. elegans. Cell Host Microbe 2009, 6, 321–330. [Google Scholar] [CrossRef] [PubMed]
  17. Keshet, A.; Mertenskötter, A.; Winter, S.A.; Brinkmann, V.; Dölling, R.; Paul, R.J. PMK-1 p38 MAPK promotes cadmium stress resistance, the expression of SKN-1/Nrf and DAF-16 target genes, and protein biosynthesis in Caenorhabditis elegans. Mol. Genet. Genom. 2017, 292, 1341–1361. [Google Scholar] [CrossRef]
  18. Wang, S.; Tang, M.; Pei, B.; Xiao, X.; Wang, J.; Hang, H.; Wu, L. Cadmium-induced germline apoptosis in Caenorhabditis elegans: The roles of HUS1, p53, and MAPK signaling pathways. Toxicol. Sci. 2008, 102, 345–351. [Google Scholar] [CrossRef]
  19. Peterson, N.D.; Icso, J.D.; Salisbury, J.E.; Rodriguez, T.; Thompson, P.R.; Pukkila-Worley, R. Pathogen infection and cholesterol deficiency activate the C. elegans p38 immune pathway through a TIR-1/SARM1 phase transition. eLife 2022, 11, e74206. [Google Scholar] [CrossRef]
  20. Hoeven, R.; McCallum, K.C.; Cruz, M.R.; Garsin, D.A. Ce-Duox1/BLI-3 generated reactive oxygen species trigger protective SKN-1 activity via p38 MAPK signaling during infection in C. elegans. PLoS Pathog. 2011, 7, e1002453. [Google Scholar] [CrossRef]
  21. Inoue, H.; Hisamoto, N.; An, J.H.; Oliveira, R.P.; Nishida, E.; Blackwell, T.K.; Matsumoto, K. The C. elegans p38 MAPK pathway regulates nuclear localization of the transcription factor SKN-1 in oxidative stress response. Genes Dev. 2005, 19, 2278–2283. [Google Scholar] [CrossRef] [PubMed]
  22. Hwang, M.; Shrestha, C.; Kang, S.; Kim, J. MEKK-3 Acts Cooperatively with NSY-1 in SKN-1-Dependent Manner against Oxidative Stress and Aging in Caenorhabditis elegans. Biology 2022, 11, 1526. [Google Scholar] [CrossRef] [PubMed]
  23. Mertenskotter, A.; Keshet, A.; Gerke, P.; Paul, R.J. The p38 MAPK PMK-1 shows heat-induced nuclear translocation, supports chaperone expression, and affects the heat tolerance of Caenorhabditis elegans. Cell Stress Chaperones 2013, 18, 293–306. [Google Scholar] [CrossRef] [PubMed]
  24. Munkacsy, E.; Khan, M.H.; Lane, R.K.; Borror, M.B.; Park, J.H.; Bokov, A.F.; Fisher, A.L.; Link, C.D.; Rea, S.L. DLK-1, SEK-3 and PMK-3 Are Required for the Life Extension Induced by Mitochondrial Bioenergetic Disruption in C. elegans. PLoS Genet. 2016, 12, e1006133. [Google Scholar] [CrossRef]
  25. Nargund, A.M.; Pellegrino, M.W.; Fiorese, C.J.; Baker, B.M.; Haynes, C.M. Mitochondrial import efficiency of ATFS-1 regulates mitochondrial UPR activation. Science 2012, 337, 587–590. [Google Scholar] [CrossRef]
  26. Nargund, A.M.; Fiorese, C.J.; Pellegrino, M.W.; Deng, P.; Haynes, C.M. Mitochondrial and nuclear accumulation of the transcription factor ATFS-1 promotes OXPHOS recovery during the UPR(mt). Mol. Cell 2015, 58, 123–133. [Google Scholar] [CrossRef] [PubMed]
  27. Ma, H.; Chen, W.; Fan, W.; He, H.; Huang, F. Roles of LonP1 in Oral-Maxillofacial Developmental Defects and Tumors: A Novel Insight. Int. J. Mol. Sci. 2022, 23, 13370. [Google Scholar] [CrossRef]
  28. Pareek, G. AAA+ proteases: The first line of defense against mitochondrial damage. PeerJ 2022, 10, e14350. [Google Scholar] [CrossRef]
  29. Pinti, M.; Gibellini, L.; Liu, Y.; Xu, S.; Lu, B.; Cossarizza, A. Mitochondrial Lon protease at the crossroads of oxidative stress, ageing and cancer. Cell. Mol. Life Sci. 2015, 72, 4807–4824. [Google Scholar] [CrossRef]
  30. Taouktsi, E.; Kyriakou, E.; Smyrniotis, S.; Borbolis, F.; Bondi, L.; Avgeris, S.; Trigazis, E.; Rigas, S.; Voutsinas, G.E.; Syntichaki, P. Organismal and Cellular Stress Responses upon Disruption of Mitochondrial Lonp1 Protease. Cells 2022, 11, 1363. [Google Scholar] [CrossRef]
  31. Hurd, D.D. Tubulins in C. elegans. WormBook 2018, 2018, 1–32. [Google Scholar] [CrossRef] [PubMed]
  32. Haynes, C.M.; Petrova, K.; Benedetti, C.; Yang, Y.; Ron, D. ClpP mediates activation of a mitochondrial unfolded protein response in C. elegans. Dev. Cell 2007, 13, 467–480. [Google Scholar] [CrossRef] [PubMed]
  33. Li, T.Y.; Sleiman, M.B.; Li, H.; Gao, A.W.; Mottis, A.; Bachmann, A.M.; El Alam, G.; Li, X.; Goeminne, L.J.E.; Schoonjans, K.; et al. The transcriptional coactivator CBP/p300 is an evolutionarily conserved node that promotes longevity in response to mitochondrial stress. Nat. Aging 2021, 1, 165–178. [Google Scholar] [CrossRef] [PubMed]
  34. Wu, X.; Li, L.; Jiang, H. Doa1 targets ubiquitinated substrates for mitochondria-associated degradation. J. Cell Biol. 2016, 213, 49–63. [Google Scholar] [CrossRef] [PubMed]
  35. Li, S.; Armstrong, C.M.; Bertin, N.; Ge, H.; Milstein, S.; Boxem, M.; Vidalain, P.-O.; Han, J.-D.J.; Chesneau, A.; Hao, T.; et al. A Map of the Interactome Network of the Metazoan C. elegans. Science 2004, 303, 540–543. [Google Scholar] [CrossRef] [PubMed]
  36. Zhong, W.; Sternberg, P.W. Genome-Wide Prediction of C. elegans Genetic Interactions. Science 2006, 311, 1481–1484. [Google Scholar] [CrossRef] [PubMed]
  37. Koyuncu, S.; Loureiro, R.; Lee, H.J.; Wagle, P.; Krueger, M.; Vilchez, D. Rewiring of the ubiquitinated proteome determines ageing in C. elegans. Nature 2021, 596, 285–290. [Google Scholar] [CrossRef] [PubMed]
  38. Heo, J.-M.; Livnat-Levanon, N.; Taylor, E.B.; Jones, K.T.; Dephoure, N.; Ring, J.; Xie, J.; Brodsky, J.L.; Madeo, F.; Gygi, S.P.; et al. A Stress-Responsive System for Mitochondrial Protein Degradation. Mol. Cell 2010, 40, 465–480. [Google Scholar] [CrossRef]
  39. Hirsch, C.; Gauss, R.; Horn, S.C.; Neuber, O.; Sommer, T. The ubiquitylation machinery of the endoplasmic reticulum. Nature 2009, 458, 453–460. [Google Scholar] [CrossRef]
  40. Xu, S.; Peng, G.; Wang, Y.; Fang, S.; Karbowski, M. The AAA-ATPase p97 is essential for outer mitochondrial membrane protein turnover. Mol. Biol. Cell 2011, 22, 291–300. [Google Scholar] [CrossRef]
  41. Yamanaka, K.; Okubo, Y.; Suzaki, T.; Ogura, T. Analysis of the two p97/VCP/Cdc48p proteins of Caenorhabditis elegans and their suppression of polyglutamine-induced protein aggregation. J. Struct. Biol. 2004, 146, 242–250. [Google Scholar] [CrossRef] [PubMed]
  42. Dunbar, T.L.; Yan, Z.; Balla, K.M.; Smelkinson, M.G.; Troemel, E.R. C. elegans detects pathogen-induced translational inhibition to activate immune signaling. Cell Host Microbe 2012, 11, 375–386. [Google Scholar] [CrossRef] [PubMed]
  43. Melo, J.A.; Ruvkun, G. Inactivation of conserved C. elegans genes engages pathogen- and xenobiotic-associated defenses. Cell 2012, 149, 452–466. [Google Scholar] [CrossRef] [PubMed]
  44. Pellegrino, M.W.; Nargund, A.M.; Kirienko, N.V.; Gillis, R.; Fiorese, C.J.; Haynes, C.M. Mitochondrial UPR-regulated innate immunity provides resistance to pathogen infection. Nature 2014, 516, 414–417. [Google Scholar] [CrossRef] [PubMed]
  45. Estes, K.A.; Dunbar, T.L.; Powell, J.R.; Ausubel, F.M.; Troemel, E.R. bZIP transcription factor zip-2 mediates an early response to Pseudomonas aeruginosa infection in Caenorhabditis elegans. Proc. Natl. Acad. Sci. USA 2010, 107, 2153–2158. [Google Scholar] [CrossRef] [PubMed]
  46. McEwan, D.L.; Kirienko, N.V.; Ausubel, F.M. Host translational inhibition by Pseudomonas aeruginosa Exotoxin A Triggers an immune response in Caenorhabditis elegans. Cell Host Microbe 2012, 11, 364–374. [Google Scholar] [CrossRef]
  47. Kirienko, N.V.; Ausubel, F.M.; Ruvkun, G. Mitophagy confers resistance to siderophore-mediated killing by Pseudomonas aeruginosa. Proc. Natl. Acad. Sci. USA 2015, 112, 1821–1826. [Google Scholar] [CrossRef]
  48. Tjahjono, E.; Kirienko, N.V. A conserved mitochondrial surveillance pathway is required for defense against Pseudomonas aeruginosa. PLoS Genet. 2017, 13, e1006876. [Google Scholar] [CrossRef]
  49. Hahm, J.H.; Jeong, C.; Lee, W.; Koo, H.J.; Kim, S.; Hwang, D.; Nam, H.G. A cellular surveillance and defense system that delays aging phenotypes in C. elegans. Aging 2020, 12, 8202–8220. [Google Scholar] [CrossRef]
  50. Strauss, K.A.; Jinks, R.N.; Puffenberger, E.G.; Venkatesh, S.; Singh, K.; Cheng, I.; Mikita, N.; Thilagavathi, J.; Lee, J.; Sarafianos, S.; et al. CODAS Syndrome Is Associated with Mutations of LONP1, Encoding Mitochondrial AAA+ Lon Protease. Am. J. Hum. Genet. 2015, 96, 121–135. [Google Scholar] [CrossRef]
  51. Zanini, G.; Selleri, V.; Malerba, M.; Solodka, K.; Sinigaglia, G.; Nasi, M.; Mattioli, A.V.; Pinti, M. The Role of Lonp1 on Mitochondrial Functions during Cardiovascular and Muscular Diseases. Antioxidants 2023, 12, 598. [Google Scholar] [CrossRef] [PubMed]
  52. Zhao, K.; Huang, X.; Zhao, W.; Lu, B.; Yang, Z. LONP1-mediated mitochondrial quality control safeguards metabolic shifts in heart development. Development 2022, 149, dev200458. [Google Scholar] [CrossRef] [PubMed]
  53. De Gaetano, A.; Gibellini, L.; Bianchini, E.; Borella, R.; De Biasi, S.; Nasi, M.; Boraldi, F.; Cossarizza, A.; Pinti, M. Impaired Mitochondrial Morphology and Functionality in Lonp1wt/− Mice. J. Clin. Med. 2020, 9, 1783. [Google Scholar] [CrossRef] [PubMed]
  54. Pareek, G.; Thomas, R.E.; Vincow, E.S.; Morris, D.R.; Pallanck, L.J. Lon protease inactivation in Drosophila causes unfolded protein stress and inhibition of mitochondrial translation. Cell Death Discov. 2018, 4, 51. [Google Scholar] [CrossRef] [PubMed]
  55. Quirós, P.M.; Español, Y.; Acín-Pérez, R.; Rodríguez, F.; Bárcena, C.; Watanabe, K.; Calvo, E.; Loureiro, M.; Fernández-García, M.S.; Fueyo, A.; et al. ATP-Dependent Lon Protease Controls Tumor Bioenergetics by Reprogramming Mitochondrial Activity. Cell Rep. 2014, 8, 542–556. [Google Scholar] [CrossRef] [PubMed]
  56. Sun, C.-L.; Van Gilst, M.; Crowder, C.M. Hypoxia-induced mitochondrial stress granules. Cell Death Dis. 2023, 14, 448. [Google Scholar] [CrossRef] [PubMed]
  57. Bounoutas, A.; Kratz, J.; Emtage, L.; Ma, C.; Nguyen, K.C.; Chalfie, M. Microtubule depolymerization in Caenorhabditis elegans touch receptor neurons reduces gene expression through a p38 MAPK pathway. Proc. Natl. Acad. Sci. USA 2011, 108, 3982–3987. [Google Scholar] [CrossRef]
  58. van der Vaart, A.; Rademakers, S.; Jansen, G. DLK-1/p38 MAP Kinase Signaling Controls Cilium Length by Regulating RAB-5 Mediated Endocytosis in Caenorhabditis elegans. PLoS Genet. 2015, 11, e1005733. [Google Scholar] [CrossRef]
  59. Butler, J.A.; Ventura, N.; Johnson, T.E.; Rea, S.L. Long-lived mitochondrial (Mit) mutants of Caenorhabditis elegans utilize a novel metabolism. FASEB J. 2010, 24, 4977–4988. [Google Scholar] [CrossRef]
  60. Hahm, J.H.; Jeong, C.; Nam, H.G. Diet restriction-induced healthy aging is mediated through the immune signaling component ZIP-2 in Caenorhabditis elegans. Aging Cell 2019, 18, e12982. [Google Scholar] [CrossRef]
  61. Hahm, J.H.; Nirmala, F.S.; Choi, P.G.; Seo, H.D.; Ha, T.Y.; Jung, C.H.; Ahn, J. The innate immune signaling component FBXC-58 mediates dietary restriction effects on healthy aging in Caenorhabditis elegans. Aging 2023, 15, 21–36. [Google Scholar] [CrossRef] [PubMed]
  62. Radetskaya, O.; Lane, R.K.; Friedman, T.; Garrett, A.; Nguyen, M.; Borror, M.B.; Russell, J.; Rea, S.L. The PMK-3 (p38) Mitochondrial Retrograde Response Functions in Intestinal Cells to Extend Life via the ESCRT Machinery. bioRxiv 2019, 797308. [Google Scholar] [CrossRef]
  63. Dayalan Naidu, S.; Sutherland, C.; Zhang, Y.; Risco, A.; de la Vega, L.; Caunt, C.J.; Hastie, C.J.; Lamont, D.J.; Torrente, L.; Chowdhry, S.; et al. Heat Shock Factor 1 Is a Substrate for p38 Mitogen-Activated Protein Kinases. Mol. Cell. Biol. 2016, 36, 2403–2417. [Google Scholar] [CrossRef] [PubMed]
  64. Kyriakou, E.; Taouktsi, E.; Syntichaki, P. The Thermal Stress Coping Network of the Nematode Caenorhabditis elegans. Int. J. Mol. Sci. 2022, 23, 14907. [Google Scholar] [CrossRef] [PubMed]
  65. Gibellini, L.; Borella, R.; De Gaetano, A.; Zanini, G.; Tartaro, D.L.; Carnevale, G.; Beretti, F.; Losi, L.; De Biasi, S.; Nasi, M.; et al. Evidence for mitochondrial Lonp1 expression in the nucleus. Sci. Rep. 2022, 12, 10877. [Google Scholar] [CrossRef] [PubMed]
  66. Melamed Kadosh, D.; Beenstock, J.; Engelberg, D.; Admon, A. Differential Modulation of the Phosphoproteome by the MAP Kinases Isoforms p38&alpha; and p38&beta. Int. J. Mol. Sci. 2023, 24, 12442. [Google Scholar] [CrossRef]
  67. Schindelin, J.; Arganda-Carreras, I.; Frise, E.; Kaynig, V.; Longair, M.; Pietzsch, T.; Preibisch, S.; Rueden, C.; Saalfeld, S.; Schmid, B.; et al. Fiji: An open-source platform for biological-image analysis. Nat. Methods 2012, 9, 676–682. [Google Scholar] [CrossRef]
Figure 1. Activation of the mitochondrial MAPK (MAPKmt) signaling pathway in lonp-1-deficient worms. (A) Schematic representation of the MAPKmt pathway consisting of the DLK-1/SEK-3/PMK-3 kinase cascade and the downstream target gene, tbb-6. (B) Relative mRNA quantification of endogenous tbb-6 gene expression levels in wt and lonp-1-knockout (ko) mutants. (C) Representative microscopy images and GFP fluorescence quantification of the tbb-6p::gfp reporter in wt and lonp-1(ko) worms. (D,E) Quantification of endogenous tbb-6 gene expression or the tbb-6p::gfp reporter in wt and lonp-1(ko) mutants upon disruption of the MAPKmt signaling components. Experiments were performed on 1-day-old adult worms grown on NGM plates, seeded with E. coli OP50 or with E. coli HT115 bacteria transformed with the indicated RNAi construct (and empty vector as a control) at 20 °C. Scale bar indicates 100 µm. Values are presented as means ± SEM, and asterisks denote statistical significance assessed with an unpaired Student’s t-test or with a two-way ANOVA followed by a post hoc Tukey’s test, ** p ≤ 0.01, *** p ≤ 0.001, **** p ≤ 0.0001.
Figure 1. Activation of the mitochondrial MAPK (MAPKmt) signaling pathway in lonp-1-deficient worms. (A) Schematic representation of the MAPKmt pathway consisting of the DLK-1/SEK-3/PMK-3 kinase cascade and the downstream target gene, tbb-6. (B) Relative mRNA quantification of endogenous tbb-6 gene expression levels in wt and lonp-1-knockout (ko) mutants. (C) Representative microscopy images and GFP fluorescence quantification of the tbb-6p::gfp reporter in wt and lonp-1(ko) worms. (D,E) Quantification of endogenous tbb-6 gene expression or the tbb-6p::gfp reporter in wt and lonp-1(ko) mutants upon disruption of the MAPKmt signaling components. Experiments were performed on 1-day-old adult worms grown on NGM plates, seeded with E. coli OP50 or with E. coli HT115 bacteria transformed with the indicated RNAi construct (and empty vector as a control) at 20 °C. Scale bar indicates 100 µm. Values are presented as means ± SEM, and asterisks denote statistical significance assessed with an unpaired Student’s t-test or with a two-way ANOVA followed by a post hoc Tukey’s test, ** p ≤ 0.01, *** p ≤ 0.001, **** p ≤ 0.0001.
Ijms 24 17209 g001
Figure 2. ATFS-1 inhibits activation of the MAPKmt pathway upstream of PMK-3. (A) Relative mRNA quantification of endogenous tbb-6 gene expression levels in wt and atfs-1(gk3094) loss-of-function mutants, or in wt and lonp-1(ko) animals subjected to RNAi against atfs-1. (B) Representative microscopy images and GFP fluorescence quantification of the tbb-6p::gfp reporter in wt, lonp-1(ko), pmk-3(ok169) and lonp-1(ko);pmk-3(ok169) worms subjected to atfs-1(RNAi). Experiments were performed on 1-day-old adult worms grown on NGM plates seeded with E. coli OP50 or with E. coli HT115 bacteria transformed with the indicated RNAi construct (and empty vector as a control) at 20 °C. Scale bar indicates 100 µm. Values are presented as means ± SEM, and asterisks denote statistical significance assessed with an unpaired Student’s t-test or with a two-way ANOVA followed by a post hoc Tukey’s test, ** p ≤ 0.01, **** p ≤ 0.0001.
Figure 2. ATFS-1 inhibits activation of the MAPKmt pathway upstream of PMK-3. (A) Relative mRNA quantification of endogenous tbb-6 gene expression levels in wt and atfs-1(gk3094) loss-of-function mutants, or in wt and lonp-1(ko) animals subjected to RNAi against atfs-1. (B) Representative microscopy images and GFP fluorescence quantification of the tbb-6p::gfp reporter in wt, lonp-1(ko), pmk-3(ok169) and lonp-1(ko);pmk-3(ok169) worms subjected to atfs-1(RNAi). Experiments were performed on 1-day-old adult worms grown on NGM plates seeded with E. coli OP50 or with E. coli HT115 bacteria transformed with the indicated RNAi construct (and empty vector as a control) at 20 °C. Scale bar indicates 100 µm. Values are presented as means ± SEM, and asterisks denote statistical significance assessed with an unpaired Student’s t-test or with a two-way ANOVA followed by a post hoc Tukey’s test, ** p ≤ 0.01, **** p ≤ 0.0001.
Ijms 24 17209 g002
Figure 3. CBP-3 positively regulates MAPKmt signaling and ATFS-1 suppresses cbp-3 induction in lonp-1 mutants. (A) Representative microscopy images and GFP fluorescence quantification of the tbb-6p::gfp reporter in wt and lonp-1(ko) worms subjected to RNAi against cbp-3. (B) Relative mRNA quantification of endogenous tbb-6 gene expression levels in wt and lonp-1(ko) worms subjected to RNAi against cbp-3. (C) Relative mRNA quantification of cbp-3 and cbp-1 gene expression levels in wt, lonp-1(ko), pmk-3(ok169) and lonp-1(ko);pmk-3(ok169) worms. (D) Relative mRNA quantification of cbp-3 gene expression levels in wt and lonp-1(ko) animals subjected to RNAi against atfs-1 or skn-1. Experiments were performed on 1-day-old adult worms grown on NGM plates seeded with E. coli OP50 or with E. coli HT115 bacteria transformed with the indicated RNAi construct (and empty vector as control) at 20 °C. Scale bar indicates 100 µm. Values are presented as means ± SEM, and asterisks denote statistical significance assessed with a two-way ANOVA followed by post hoc Tukey’s test, * p ≤ 0.05, ** p ≤ 0.01, **** p ≤ 0.0001.
Figure 3. CBP-3 positively regulates MAPKmt signaling and ATFS-1 suppresses cbp-3 induction in lonp-1 mutants. (A) Representative microscopy images and GFP fluorescence quantification of the tbb-6p::gfp reporter in wt and lonp-1(ko) worms subjected to RNAi against cbp-3. (B) Relative mRNA quantification of endogenous tbb-6 gene expression levels in wt and lonp-1(ko) worms subjected to RNAi against cbp-3. (C) Relative mRNA quantification of cbp-3 and cbp-1 gene expression levels in wt, lonp-1(ko), pmk-3(ok169) and lonp-1(ko);pmk-3(ok169) worms. (D) Relative mRNA quantification of cbp-3 gene expression levels in wt and lonp-1(ko) animals subjected to RNAi against atfs-1 or skn-1. Experiments were performed on 1-day-old adult worms grown on NGM plates seeded with E. coli OP50 or with E. coli HT115 bacteria transformed with the indicated RNAi construct (and empty vector as control) at 20 °C. Scale bar indicates 100 µm. Values are presented as means ± SEM, and asterisks denote statistical significance assessed with a two-way ANOVA followed by post hoc Tukey’s test, * p ≤ 0.05, ** p ≤ 0.01, **** p ≤ 0.0001.
Ijms 24 17209 g003
Figure 4. The MAD pathway does not mediate activation of the MAPKmt pathway in lonp-1 mutants. (A) Representative microscopy images and GFP fluorescence quantification of the tbb-6p::gfp reporter in 1-day adults of wt, cdc-48.2(tm659), lonp-1(ko) and lonp-1(ko);cdc-48.2(tm659), at 20 °C. (B) Relative mRNA quantification of endogenous tbb-6 gene expression levels in 1-day adults of the aforementioned strains, grown on NGM plates seeded with E. coli OP50 or with E. coli HT115 bacteria transformed with the cdc-48.1(RNAi) construct (and empty vector as control), at 20 °C. Scale bar indicate 100 µm. Values were presented as mean ± SEM, and numeric data were analyzed with a two-way ANOVA followed by post hoc Tukey’s test, showing no significant (ns) differences.
Figure 4. The MAD pathway does not mediate activation of the MAPKmt pathway in lonp-1 mutants. (A) Representative microscopy images and GFP fluorescence quantification of the tbb-6p::gfp reporter in 1-day adults of wt, cdc-48.2(tm659), lonp-1(ko) and lonp-1(ko);cdc-48.2(tm659), at 20 °C. (B) Relative mRNA quantification of endogenous tbb-6 gene expression levels in 1-day adults of the aforementioned strains, grown on NGM plates seeded with E. coli OP50 or with E. coli HT115 bacteria transformed with the cdc-48.1(RNAi) construct (and empty vector as control), at 20 °C. Scale bar indicate 100 µm. Values were presented as mean ± SEM, and numeric data were analyzed with a two-way ANOVA followed by post hoc Tukey’s test, showing no significant (ns) differences.
Ijms 24 17209 g004
Figure 5. PMK-3-dependent activation of the bZIP transcription factor ZIP-2 does not confer resistance to pathogen infection. (A) Relative mRNA quantification of zip-2, irg-1 and irg-2 gene expression levels in 1-day-old adults of wt, lonp-1(ko), zip-2(ok3730), lonp-1(ko);zip-2(ok3730), pmk-3(ok169) and lonp-1(ko);pmk-3(ok169) at 20 °C. Values are presented as means ± SEM and statistical significance was assessed with an unpaired Student’s t-test, * p ≤ 0.05, ** p ≤ 0.01, *** p ≤ 0.001 and **** p ≤ 0.0001. (B) Survival of slow killing Pseudomonas aeruginosa PA14 pathogen infection assay in 1-day-old adults of wt, lonp-1(ko), pmk-3(ok160) and lonp-1(ko);pmk-3(ok169) on NGM plates seeded with E. coli OP50 in the presence of FUdR at 20 °C. Statistical significance was assessed via a Kaplan–Meier survival curve, **** p ≤ 0.0001.
Figure 5. PMK-3-dependent activation of the bZIP transcription factor ZIP-2 does not confer resistance to pathogen infection. (A) Relative mRNA quantification of zip-2, irg-1 and irg-2 gene expression levels in 1-day-old adults of wt, lonp-1(ko), zip-2(ok3730), lonp-1(ko);zip-2(ok3730), pmk-3(ok169) and lonp-1(ko);pmk-3(ok169) at 20 °C. Values are presented as means ± SEM and statistical significance was assessed with an unpaired Student’s t-test, * p ≤ 0.05, ** p ≤ 0.01, *** p ≤ 0.001 and **** p ≤ 0.0001. (B) Survival of slow killing Pseudomonas aeruginosa PA14 pathogen infection assay in 1-day-old adults of wt, lonp-1(ko), pmk-3(ok160) and lonp-1(ko);pmk-3(ok169) on NGM plates seeded with E. coli OP50 in the presence of FUdR at 20 °C. Statistical significance was assessed via a Kaplan–Meier survival curve, **** p ≤ 0.0001.
Ijms 24 17209 g005
Figure 6. Loss of ZIP-2 or PMK-3 shortened the lifespan of lonp-1 mutants and PMK-3 promotes organismal survival under stress. (A) Lifespan assays of wt, lonp-1(ko), zip-2(ok3730), lonp-1(ko);zip-2(ok3730), pmk-3(ok169) and lonp-1(ko);pmk-3(ok169) worms on NGM plates seeded with E. coli OP50 at 20 °C. Replicates and statistical analysis of lifespan assays are shown in Table S3, ** p ≤ 0.01, *** p ≤ 0.001. (B) Survival of 1-day-old adults of wt, lonp-1(ko), pmk-3 ok160) and lonp-1(ko);pmk-3(ok160) strains treated with tert-butyl hydroperoxide (tBHP, 10 mM), Antimycin A (40 μm for 24 h), or subjected to heat shock (at 35 °C for 5.5, 7.5 or 8 h). The percentage survival for all biological replicates was plotted, and an unpaired Student’s t-test was used to assess significance, * p ≤ 0.05, ** p ≤ 0.01 and **** p ≤ 0.0001.
Figure 6. Loss of ZIP-2 or PMK-3 shortened the lifespan of lonp-1 mutants and PMK-3 promotes organismal survival under stress. (A) Lifespan assays of wt, lonp-1(ko), zip-2(ok3730), lonp-1(ko);zip-2(ok3730), pmk-3(ok169) and lonp-1(ko);pmk-3(ok169) worms on NGM plates seeded with E. coli OP50 at 20 °C. Replicates and statistical analysis of lifespan assays are shown in Table S3, ** p ≤ 0.01, *** p ≤ 0.001. (B) Survival of 1-day-old adults of wt, lonp-1(ko), pmk-3 ok160) and lonp-1(ko);pmk-3(ok160) strains treated with tert-butyl hydroperoxide (tBHP, 10 mM), Antimycin A (40 μm for 24 h), or subjected to heat shock (at 35 °C for 5.5, 7.5 or 8 h). The percentage survival for all biological replicates was plotted, and an unpaired Student’s t-test was used to assess significance, * p ≤ 0.05, ** p ≤ 0.01 and **** p ≤ 0.0001.
Ijms 24 17209 g006
Figure 7. A schematic representation of the genetic interactions and molecular functions of MAPKmt in worms deficient in the lonp-1 gene.
Figure 7. A schematic representation of the genetic interactions and molecular functions of MAPKmt in worms deficient in the lonp-1 gene.
Ijms 24 17209 g007
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

Taouktsi, E.; Kyriakou, E.; Voulgaraki, E.; Verganelakis, D.; Krokou, S.; Rigas, S.; Voutsinas, G.E.; Syntichaki, P. Mitochondrial p38 Mitogen-Activated Protein Kinase: Insights into Its Regulation of and Role in LONP1-Deficient Nematodes. Int. J. Mol. Sci. 2023, 24, 17209. https://doi.org/10.3390/ijms242417209

AMA Style

Taouktsi E, Kyriakou E, Voulgaraki E, Verganelakis D, Krokou S, Rigas S, Voutsinas GE, Syntichaki P. Mitochondrial p38 Mitogen-Activated Protein Kinase: Insights into Its Regulation of and Role in LONP1-Deficient Nematodes. International Journal of Molecular Sciences. 2023; 24(24):17209. https://doi.org/10.3390/ijms242417209

Chicago/Turabian Style

Taouktsi, Eirini, Eleni Kyriakou, Evangelia Voulgaraki, Dimitris Verganelakis, Stefania Krokou, Stamatis Rigas, Gerassimos E. Voutsinas, and Popi Syntichaki. 2023. "Mitochondrial p38 Mitogen-Activated Protein Kinase: Insights into Its Regulation of and Role in LONP1-Deficient Nematodes" International Journal of Molecular Sciences 24, no. 24: 17209. https://doi.org/10.3390/ijms242417209

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