[go: up one dir, main page]

Skip to main content
Molecular and Cellular Biology logoLink to Molecular and Cellular Biology
. 2023 Oct 8;43(10):531–546. doi: 10.1080/10985549.2023.2253131

Mitochondrial Fragmentation Promotes Inflammation Resolution Responses in Macrophages via Histone Lactylation

Leah I Susser a,b, My-Anh Nguyen a,b, Michele Geoffrion b, Christina Emerton b, Mireille Ouimet a,b, Mireille Khacho a,c,d, Katey J Rayner a,c,b,
PMCID: PMC10569354  PMID: 37807652

Abstract

During the inflammatory response, macrophage phenotypes can be broadly classified as pro-inflammatory/classically activated “M1”, or pro-resolving/alternatively “M2” macrophages. Although the classification of macrophages is general and assumes there are distinct phenotypes, in reality macrophages exist across a spectrum and must transform from a pro-inflammatory state to a proresolving state following an inflammatory insult. To adapt to changing metabolic needs of the cell, mitochondria undergo fusion and fission, which have important implications for cell fate and function. We hypothesized that mitochondrial fission and fusion directly contribute to macrophage function during the pro-inflammatory and proresolving phases. In the present study, we find that mitochondrial length directly contributes to macrophage phenotype, primarily during the transition from a pro-inflammatory to a proresolving state. Phenocopying the elongated mitochondrial network (by disabling the fission machinery using siRNA) leads to a baseline reduction in the inflammatory marker IL-1β, but a normal inflammatory response to LPS, similar to control macrophages. In contrast, in macrophages with a phenocopied fragmented phenotype (by disabling the fusion machinery using siRNA) there is a heightened inflammatory response to LPS and increased signaling through the ATF4/c-Jun transcriptional axis compared to control macrophages. Importantly, macrophages with a fragmented mitochondrial phenotype show increased expression of proresolving mediator arginase 1 and increased phagocytic capacity. Promoting mitochondrial fragmentation caused an increase in cellular lactate, and an increase in histone lactylation which caused an increase in arginase 1 expression. These studies demonstrate that a fragmented mitochondrial phenotype is critical for the proresolving response in macrophages and specifically drive epigenetic changes via lactylation of histones following an inflammatory insult.

Keywords: mitochondrial metabolism, macrophages, fission, fusion, inflammation resolution, histone lactylation

GRAPHICAL ABSTRACT

graphic file with name TMCB_A_2253131_UF0001_C.jpg

INTRODUCTION

Macrophages are a major component of the innate immune response, where they sense their external environment and adopt specific phenotypes according to the cues received. Macrophage phenotypes can be broadly classified into M1 or M2, with M1 macrophages being pro-inflammatory and M2 macrophages being proresolving or anti-inflammatory. M1 macrophages produce high levels of pro-inflammatory cytokines (e.g. TNFα and IL1β), reactive oxygen species and promote Th1 responses. In contrast, M2 macrophages secrete anti-inflammatory cytokines, have high efferocytic capacity and produce collagen and other proteins to enable tissue repair.1–3 The spectrum of M1 to M2 is dynamic and necessary for the appropriate response to injury or infection. However, the imbalance of these two phenotypes can alter the progression and outcome of numerous diseases, such as cancer and atherosclerosis.4–6 It is, therefore, important to decipher the factors that control macrophage polarization to better understand normal and pathophysiological processes.

Macrophages have distinct metabolic needs that enable them to adapt to tissue surroundings and functional demands. The metabolic differences between M1 and M2 macrophages have been extensively characterized, where M1 macrophages preferentially use glycolytic metabolism, while M2 macrophages primarily use oxidative metabolism to generate ATP.7 Beyond ATP generation, macrophages use metabolic signals derived from mitochondria to trigger other functions in the inflammatory response: acetyl-coA is used to acetylate histones to promote inflammatory gene expression, succinate accumulation stabilizes HIF1α and α-ketoglutarate is used to demethylate specific anti-inflammatory genes.8 Mitochondria are also part of a communicating network that regularly undergo morphological changes to adapt to the metabolic needs of the cell.9 Mitochondrial dynamics are controlled by opposing functions of fission and fusion. Fission is primarily carried out through the action of cytosolic dynamin-related protein 1 (Drp1) and two outer mitochondrial membrane-bound proteins, mitochondrial fission factor (Mff) and mitochondrial fission 1 (Fis1). Fusion is controlled mainly through the action of outer membrane-bound mitofusin 1/2 (Mfn 1 & Mfn2) and inner membrane-bound optic atrophy 1 (Opa1).10

Mitochondrial dynamics play a critical role in cell health and function. Mutations in all these proteins are fetal-lethal in mice and lead to devastating neurodegenerative diseases in humans.11,12 Fission and fusion have also been shown to play a directional role in cell differentiation, including T cell phenotype commitment and neuronal and muscle stem cell differentiation.13,14 Individual components of the fission or fusion machinery (i.e. Drp1 or Opa1) have been linked to pro-inflammatory macrophage responses,15–18 however, little is known about whether mitochondrial dynamism and the action of fission and fusion control pro- or anti-inflammatory macrophage phenotypes. In the present study, we determine the role of fission and fusion on pro and anti-inflammatory pathways and find that a fragmented mitochondrial phenotype directly contributes to proresolving macrophage function following inflammatory activation.

RESULTS

Macrophage polarization influences mitochondrial fission and fusion

To understand how macrophage polarization affects mitochondrial morphology, we first characterized the mitochondrial network of bone marrow-derived macrophages (BMDMs) polarized to traditional M1 or M2 phenotypes. We observed that macrophages stimulated with LPS and IFNγ for 24 h (pro-inflammatory or M1) have a significantly longer mitochondrial network compared to both unpolarized macrophages (resting, M0), and IL-4 stimulated macrophages (proresolving or M2) (Fig. 1a, b, d, Supplemental Movies a-c). The increase in mitochondrial length was observed as early as 2 h after stimulation and prior to the establishment of an M1 identity (Fig. 1e and f). Mitochondrial morphology and changes in mitochondrial length between M0, M1 and M2 macrophages was confirmed by transmission electron microscopy showing longer mitochondria in M1, and a higher ratio of mitochondria undergoing dynamic events (either fission or fusion) (Fig. 1g to i). Furthermore, we saw an increase in cristae width in M1 macrophages compared to M0 and M2 (Fig. 1j). In human THP-1 macrophages polarized to M1 with LPS + IFNγ, there was a similar increase in mitochondrial length compared to both M0 and M2 (Fig. 1a and c). These data demonstrate that pro-inflammatory and M1 macrophages adopt an elongated mitochondrial network, whereas resting and M2 macrophages adopt a shorter mitochondrial network that corresponds with their polarized phenotype.

FIG 1.

FIG 1

Elongated mitochondria are associated with a pro-inflammatory macrophage phenotype. BMDM or THP1 cells were differentiated and then polarized for 24 h before being fixed and analyzed by microscopy. (a) Representative immunofluorescence (IF) microscopy of polarized macrophages mitochondrial network (Tom20 in red and DAPI in blue). (b) Quantification of BMDM mitochondrial length measured manually on ImageJ (n = 3). (c) Quantification of THP1 mitochondrial length measured manually on ImageJ (n = 3). (d) Distribution of mitochondrial length across polarization (n = 3). (e) Quantification of mitochondrial length over time (n = 3). (f) Gene expression of M1 and M2 markers over time (n = 10). (g) Representative transmission electron microscopy (TEM) images of M0, M1 and M2 BMDMs. (h) Quantification of average mitochondrial length (n = 10). (i) Ratio of dynamic over static mitochondria with dynamic events defined as membrane pinching examples indicated by arrows (n = 10). (j) Quantification of average cristae width (n = 10). Data are presented as the mean of biological replicates ± SEM. Statistical analysis by one-way ANOVA.

Promoting mitochondrial fusion does not alter pro-inflammatory IL-1β responses in macrophages

Previous studies have demonstrated that mitochondrial fission and fusion control cell phenotype commitment (e.g. neural and muscle stem cells, T cells).13,14,19 Therefore, we next sought to determine whether mitochondrial fission and/or fusion directs macrophage M1 or M2 polarization. To phenocopy the hyperfused mitochondrial network in M1 macrophages, we developed a model wherein the mitochondrial fission machinery was silenced using a combination of siRNAs targeting Drp1, Mff and Fis1 to prevent mitochondrial fission (fission KD, Fig. 2a). Fission knockdown resulted in a significantly elongated mitochondrial network compared to control siRNA treated macrophages (Fig. 2b and c). We confirmed knockdown for each siRNA combination at both the mRNA and protein levels (Fig. 2d and e). We assessed pro- and anti-inflammatory gene expression in macrophages from fission KD cells both at baseline (unstimulated) and in response to inflammatory stimulation to LPS- a prototypical model of inflammatory activation. In unstimulated macrophages, phenocopying a hyperfused mitochondrial network resulted in a decreased expression of the M1 markers Il1b & Tnfa, whereas Nos2 expression was increased which corresponds with decreased M2 marker Arg1 (Fig. 2f and g). To determine whether macrophages with hyperfused mitochondria respond normally to an inflammatory stimulus, we assessed Il1b gene expression after LPS stimulation. Macrophages with elongated mitochondria had decreased Il1b gene expression after 24 h of LPS stimulation compared to controls (Fig. 2h). To assess how changes in gene expression may impact protein function, we examined the expression of the precursor, mature and secreted forms of IL-1β protein in response to LPS stimulation over time. The loss of mitochondrial fission had no effect on either pro-IL-1β or IL-1β secretion (Fig. 2i to k). To determine whether elongated mitochondria influenced the full activation of IL-1β protein secretion, which requires the engagement of the NLPR3 inflammasome via a priming and an activation signal, we tested an in vitro model of inflammasome activation with LPS priming followed by ATP activation.20–23 Priming with LPS and subsequent activation by ATP in macrophages with hyperfused mitochondria resulted in equivalent IL-1β release compared to controls, indicating a normal inflammasome priming and activation process (Fig. 2l). Together, these data indicate that while mitochondrial fusion appears to control the basal expression of macrophage Il1b gene expression, it has little influence on IL-1β protein secretion upon inflammatory stimulation. Therefore, although M1 macrophages adopt a lengthened mitochondrial network in parallel with their pro-inflammatory gene expression state, forcing an elongated mitochondrial network does not directly cause a heightened inflammatory response to LPS.

FIG 2.

FIG 2

Knocking down the mitochondrial fission machinery does not alter the IL-1β inflammatory pathway. BMDMs were transfected with siRNA against all three fission proteins or an equal concentration of scrambled control siRNA for 72 h. (a) Infographic of fission KD by siRNA transfection. (b) Representative IF microscopy of control vs fission KD cells mitochondrial network (Tom20 in red and DAPI in blue). (c) Quantification of average mitochondrial length per cell (n = 3). (d) Quantification of fission knockdown (KD) efficiency by qPCR (n = 9). (e) Verification of fission KD by Western blot (DRP1 27 kDa, FIS1 17 kDa, MFF 38 kDa, GAPDH 36 kDa, and HSP90 90 kDa). (f) Effect of fission KD on M1 marker gene expression (n = 5). (g) Effect of fission KD on M2 marker gene expression (n = 8). (h) Il-1b gene expression upon LPS stimulation in fission KD cells (n = 4). (i) Representative Western blot of IL-1β upon LPS stimulation (IL-1β 35, 29 kDa, β-Actin 45 kDa). (j) Quantification of fission KD effect on IL-1β protein upon LPS stimulation normalized to β-Actin (n = 5). (k) Secreted IL-1β levels in fission KD cells upon 24 h LPS stimulation measured by ELISA (n = 3). (l) Secreted IL-1β levels in fission KD cells upon 24 h LPS + 30 min ATP stimulation measured by ELISA (n = 3). Data are presented as the mean of biological replicates ± SEM. Statistical analysis by unpaired Student’s t test except for h where multiple unpaired t tests with Holm–Šídák correction were applied.

A fragmented mitochondrial network alters pro-inflammatory gene expression in response to LPS via noncanonical inflammatory pathways

Because we observed that M2-polarized macrophages have a more fragmented mitochondrial network compared to M1 (Fig. 1a and b), we sought to phenocopy this state by silencing mitochondrial fusion machinery using siRNA targeting Opa1, Mfn1 and Mfn2 (fusion KD, Fig. 3a). As expected, silencing the fusion machinery resulted in a fragmented mitochondrial network (Fig. 3b to e). Interestingly, similar to what we observed upon knocking down of the mitochondrial fission machinery, fusion KD resulted in a decrease in basal Il1b gene expression in unstimulated macrophages (Fig. 3f). However, unlike what happened on the loss of mitochondrial fission, loss of mitochondrial fusion resulted in a basal increase in the M2 polarization marker Arg1 (Fig. 3g). To determine whether preventing mitochondrial fusion would result in an impaired inflammatory response, we stimulated control and fusion KD macrophages with LPS and found that impaired mitochondrial fusion results in increased Il1b gene expression after 6 and 24 h (Fig. 3h). Activation of IL-1β protein was assessed after LPS stimulation where loss of mitochondrial fusion results in an increase in pro-IL-1β at 6 and 24 h (Fig. 3i) and a corresponding small but not statistically significant increase in IL-1β secretion at 24 h (Fig. 3j). Upon inflammasome activation with LPS and ATP (as described above), loss of mitochondrial fusion led to a significant increase in IL-1β secretion (Fig. 3j). These data are similar to what is observed in a parallel model of impaired mitochondrial fusion where genetic loss of Opa1 led to an increase in IL-1β secretion upon inflammasome activation,15 and support that this is likely due to functional losses in mitochondrial fusion.

FIG 3.

FIG 3

Impairing mitochondrial fusion augments IL-1β and ATF-4/c-Jun pathway. BMDMs were transfected with siRNA against all three fusion proteins or an equal concentration of scrambled control siRNA for 72 h. (a) Infographic of fusion KD by siRNA transfection. (b) Representative IF microscopy of control vs fusion KD cells mitochondrial network (Tom20 in red and DAPI in blue). (c) Quantification of average mitochondrial length per cell (n = 5). (d) Quantification of fusion knockdown (KD) efficiency by qPCR (n = 10). (e) Verification of fusion KD by Western blot (OPA1 86, 92 kDa, MFN1/2 75 kDa, and β-actin 45 kDa). (f) Effect of fusion KD on M1 marker gene expression (n = 5). G) Effect of fusion KD on M2 Marker gene expression (n = 5). H) Il-1b gene expression upon LPS stimulation in fusion KD cells (n = 4). (i) Quantification of fusion KD pro-IL-1β protein levels normalized to β-Actin upon 6 and 24 h of LPS stimulation (n = 5). (j) Secreted IL-1β levels in fusion KD cells upon 24 h LPS stimulation with and without 30 min ATP measured by ELISA (n = 3). (k) Quantification of phosphorylated p65 (NFκB) protein normalized to β-Actin after 24 h of LPS stimulation (n = 3). (l) HIF1α nuclear translocation as measured by IF microscopy upon LPS stimulation (n = 6.) (m) Representative Western blot of ATF4 upon LPS stimulation (ATF4 49 kDa & HSP90 90 kDa) and quantification (n = 3). n) Representative western blot of c-Jun and phosphorylated c-Jun upon LPS stimulation (c-Jun 43, 48 kDa, Phos c-Jun 48 kDa, HSP90 90 kDa) and quantification (n = 4). (o) Slc7a11 gene expression upon LPS stimulation in fusion KD cells (n = 4). Data are presented as the mean of biological replicates ± SEM. Statistical analysis by unpaired Student’s t test except for h and l-o, where multiple unpaired t tests with Holm–Šídák correction were applied.

To investigate how mitochondrial fusion alters the expression of IL-1β upon LPS stimulation, we investigated transcriptional pathways known to promote cytokine production. We first assessed NFκB activity, which is a potent inducer of Il1b expression and is quickly activated upon LPS stimulation, where the p65 subunit becomes phosphorylated and translocates to the nucleus to activate inflammatory gene expression.24–26 We assessed NFκB activity via measuring the phosphorylation of the p65 subunit by Western blot. Surprisingly, despite increases in Il1b gene expression upon loss of mitochondrial fusion, we find that p65 phosphorylation is significantly decreased after 24 h of LPS stimulation (Fig. 3k, Western blot not shown). These data indicate that the observed increase in IL-1β expression and secretion upon loss of mitochondrial fusion may be caused by an alternative NFκB-independent mechanism. HIF1α nuclear translocation, which is stimulated in response to LPS and drives inflammatory cytokine production downstream of NFκB was next assessed by microscopy.27 Upon LPS stimulation, the loss of mitochondrial fusion caused a decrease in HIF1α nuclear translocation (Fig. 3l). This observed decrease in HIF1α translocation would not be expected to result in increased IL-1β secretion upon LPS stimulation; therefore we looked into alternative pathways that are known to be downstream of inflammatory stimulation and may activate cytokine expression. We assessed the activity of the ATF4/c-Jun pathway, which can promote Il1b expression in response to LPS.28 When mitochondrial fusion is impaired, both ATF4 and phosphorylated c-Jun are significantly increased after 6 h of LPS stimulation (Fig. 3m and n). To confirm the increase in ATF4 activity upon knockdown of mitochondrial fusion, another ATF4-responsive gene, Slc7a11 was measured after both 6 h and 24 h of LPS stimulation,29 and indeed was increased in macrophages with fusion KD (Fig. 3o). Together these data indicate that, in response to inflammatory stimulation, mitochondrial fusion contributes to the macrophage inflammatory IL-1β response possibly via activation of the ATF4/c-Jun axis.

A fragmented mitochondrial network enhances the proresolving macrophage phenotype

Given that M2 macrophages had a more fragmented mitochondrial network compared to M1 macrophages (Fig. 1a and b) and that loss of mitochondrial fusion led to an increase in baseline expression of Arg1 (Fig. 3g), we sought to determine whether mitochondrial fusion alters the anti-inflammatory macrophage phenotype during the resolution phase of inflammation. Arginase 1 is a critical enzyme in macrophages that promotes the resolution of inflammation by reducing nitric oxide signaling, promoting L-arginine metabolism to assist in tissue repair.30 In normal macrophages, Arg1 expression is temporally activated by LPS, with expression appearing late following LPS stimulation, following the peak of the pro-inflammatory response.31 We therefore assessed whether a fragmented mitochondrial network influenced the late activation of Arg1. Upon LPS stimulation, Arg1 gene expression was maximally increased in mitochondrial fusion-impaired cells after 24 h, translating to an increased protein level at 48 h (Fig. 4a to c). IL-4 is a potent inducer of M2 macrophage responses and is produced by Th2 cells to trigger the anti-inflammatory program in macrophages.32,33 We therefore assessed the impact of impaired mitochondrial fusion upon direct anti-inflammatory stimulation using IL-4. Arg1 gene and protein expression were both increased upon fusion KD after 24 h of IL-4 treatment (Fig. 4d to f). To test how mitochondrial fusion impacts macrophage plasticity, we first stimulated cells with LPS for 6 h, after which we stimulated them with IL-4, to accelerate the proresolving phase. Loss of mitochondrial fusion increased Arg1 gene expression and protein compared to control cells, indicating a heightened proresolving response (Fig. 4g to i). A key function of M2 proresolving macrophages is phagocytic capacity; therefore, we measured phagocytosis of latex beads. Loss of mitochondrial fusion resulted in an increase in phagocytosis compared to controls basally and under LPS, IL-4 or combined stimulation. (Fig. 4j to m). Together these data indicate that inducing a fragmented mitochondrial phenotype promotes inflammation resolution following inflammatory or anti-inflammatory activation.

FIG 4.

FIG 4

Silencing mitochondrial fusion machinery promotes a proresolving macrophage phenotype. (a) Arg1 gene expression over time upon LPS stimulation (n = 4). (b) Representative ARG1 Western blot after 48 h of LPS stimulation. (c) Quantification of ARG1 protein levels normalized to β-Actin after 48 h LPS stimulation (n = 4). (d) Arg1 gene expression over time upon IL-4 stimulation (n = 5). (e) Representative ARG1 Western blot after 24 h of IL-4 stimulation. (f) Quantification of ARG1 protein levels normalized to HSP90 after 24 h IL-4 stimulation (n = 3). (g) Arg1 gene expression upon 6 h LPS + 18 h IL-4 stimulation (n = 2). (h) Representative ARG1 Western blot after 6 h LPS + 18 h IL-4 stimulation. (i) Quantification of ARG1 protein levels normalized to HSP90 after 6 h LPS + 18 h IL-4 stimulation (n = 3). (j) Representative immunofluorescence microscopy of bead phagocytosis (Tom20 in red and beads in green). (k) Quantification of bead phagocytosis in fusion KD cells under LPS stimulation (n = 4). (l) Quantification of bead phagocytosis in fusion KD cells under IL-4 stimulation (n = 3). (m) Quantification of bead phagocytosis in fusion KD cells under 6 h LPS + 18 h IL-4 stimulation (n = 4). Western blot molecular weights ARG1 40 kDa, β-Actin 45 kDa, HSP90 90 kDa. Data are presented as the mean of biological replicates ± SEM. Statistical analysis c, f, g, i, l, m by unpaired Student’s t test and a, d, k by multiple unpaired t tests with Holm–Šídák correction.

Mitochondrial fragmentation increases histone lactylation and promotes an M2 phenotype

Pro-inflammatory macrophages are highly glycolytic, and as a result, produce high levels of lactate upon inflammatory stimulation.34,35 Lactate is a powerful signaling molecule and is known to promote M2-like responses in tumor macrophages.36 It was recently shown that once sufficiently elevated, lactate causes a change in gene expression via lactylation of lysine residue on histones, turning on proresolving machinery such as Arg1.37–39 To understand whether mitochondrial fragmentation promotes changes in lactate metabolism, we assessed lactate levels in control and fusion KD cells upon LPS stimulation. After 24 h of LPS treatment, lactate levels were increased in fusion KD compared to controls (Fig. 5a). In agreement with the increase in lactate levels, histone lactylation (marked by the pan-histone lactylation antibody, KLA) was increased with knockdown of mitochondrial fusion after 24 h of LPS stimulation (Fig. 5b and c). Both lactate and histone lactylation were equivalent to that of control macrophages after 48 h of LPS stimulation (Fig. 5a to c). To better understand the increase in lactate and KLA upon loss of mitochondrial fusion, we first measured the expression of pyruvate dehydrogenase (PDH), which converts pyruvate to acetyl-CoA to enter the TCA cycle.35 Upon loss of mitochondrial fusion, we observe a decrease in PDH expression after 24 h of LPS stimulation (Fig. 5d and e). It would be expected that lower PDH expression in cells with fragmented mitochondria would result in reduced conversion of pyruvate to acetyl-coA, and an increase in conversion of pyruvate to lactate.40 To test this hypothesis, we inhibited lactate accumulation by inhibiting the activity of lactate dehydrogenase (LDH) using sodium oxamate (Ox).36 Co-treatment of macrophages with LPS and sodium oxamate for 48 h prevented the induction of ARG1 protein in fusion KD macrophages (Fig. 5f and g) and induction of KLA in control cells (Fig. 5h and i). In summary, the above data indicate that increased lactate accumulation in cells with fragmented mitochondria is driving the increase in Arg1 expression in macrophages following inflammatory activation.

FIG 5.

FIG 5

Silencing mitochondrial fusion machinery alters cellular lactate levels, increases histone lactylation and reduces PDH. (a) Lactate levels over time in fusion KD cells compared to control cells (n = 3). (b) Representative Western blot of histone lactylation (KLA) over time. (c) Quantification KLA in fusion KD cells after 24 and 48 h LPS stimulation (n = 3). (d) Representative Western blot of pyruvate dehydrogenase levels over time. (e) Quantification PDH in fusion KD cells after 24 and 48 h LPS stimulation (n = 3). (f) Representative Western blot of ARG1 in control vs fusion KD cells upon 48 h LPS stimulation with LDH inhibitor sodium oxamate. (g) Quantification ARG1 in fusion KD cells after 48 h LPS stimulation with sodium oxamate (n = 3). (h) Representative Western blot of KLA in control vs fusion KD cells upon 48 h LPS stimulation with sodium oxamate. (i) Quantification KLA in fusion KD cells after 48 h LPS stimulation with sodium oxamate (n = 3). j) Representative images of mitochondrial network morphology upon late-stage LPS stimulation (Tom20 in red and DAPI in blue). (k) Quantification of average mitochondrial length per cell (n = 4) (l) Average mitochondrial length distribution per time point (n = 4). (m) Lactate levels upon LPS stimulation in control cells with statistics vs 0h (n = 3). (n) Representative time course Western blot of histone lactylation (KLA) in untransfected BMDMs upon LPS stimulation and quantification (n = 3). (o) Representative Western blot of ARG1 in control cells upon 72 h LPS stimulation with sodium oxamate and quantification (n = 3). (p) Representative Western blot of KLA upon 72 h LPS stimulation with sodium oxamate and quantification (n = 3). Western blot molecular weights KLA 15 kDa, PDH 43 kDa, ARG1 40 kDa, β-Actin 45 kDa and HSP90 90 kDa. Data are presented as the mean of biological replicates ± SEM. Statistical analysis a, c, f, o, p by unpaired Student’s t test and g, i, k, m, n by one-way ANOVA.

The observation that siRNA-induced mitochondrial fragmentation resulted in an increase in histone lactylation and Arg1 expression prompted us to examine whether this occurs also during the physiological macrophage resolution response. Having observed a fragmented mitochondrial network in IL-4 treated M2 polarized macrophages, so we next sought to characterize the mitochondrial phenotype of macrophages as they enter the proresolving phase after LPS exposure, during which the switch to histone lactylation occurs and Arg1 expression is induced. Analysis of mitochondrial morphology after 24, 48, and 72 h following LPS stimulation revealed maximal mitochondrial elongation at 24 h, and a return to near baseline length after 48 and 72 h (Fig. 5j to l). There was a corresponding increase in lactate (Fig. 5m) and histone lactylation, as measured by KLA, between 24 and 72 h (Fig. 5n). Furthermore, co-treatment with LDH inhibitor sodium oxamate and LPS for 72 h prevented the induction of both ARG1 and KLA (Fig. 5o and p). Together, these data indicate that a fragmented mitochondrial network is associated with an increase in histone lactylation during the resolution phase of inflammation.

DISCUSSION

Until recently, mitochondrial fission and fusion were believed to be adaptations to meet the metabolic demands of the cell. However, more recent evidence demonstrates that mitochondrial fission and fusion produce specific signals to other organelles to dictate cell differentiation and cell phenotype. For example, neural and muscle stem cell self-renewal is maintained by an elongated mitochondrial network, which produces low ROS stimulation and activates notch signaling, whereas mitochondrial fragmentation leads to higher ROS levels, activation of NRF2 and transcriptional activation of neuronal commitment program.13,14,41 In T cells, commitment to an effector or memory cell phenotype is driven by Opa1 by controlling cristae morphology, thereby impacting metabolic potential. Effector T cells are characterized as having a fission-associated expansion of cristae leading to inefficient oxidative phosphorylation and a compensatory use of glycolysis as a primary form of metabolism.19 These studies indicate that mitochondrial dynamism is not only a consequence of the cellular environment, but rather plays a vital functional role in determining cellular phenotype and function. In this study, we find that a fragmented mitochondrial network is found in macrophages during the proresolving phase of inflammation. Mechanistically, this occurs via an accumulation of lactate, resulting in the lactylation of histones and a shift toward an anti-inflammatory phenotype. These data are the first to show that mitochondrial dynamics regulate epigenetic modification of proresolving genes in macrophages in response to inflammatory cues.

Following the pro-inflammatory response, in order to return to baseline, macrophages need to adopt an anti-inflammatory and proresolving phenotype. This requires the dampening of pro-inflammatory cytokine production, the activation of proreparative gene programs and a switch to an oxidative metabolic phenotype. Arginase 1 is a key mediator of proreparative macrophage responses, where it is turned on during the resolution phase to promote the production of proline and polyamines for wound repair and proliferation.42–44 Arg1 expression is high in M2 macrophages (i.e. upon IL-4 stimulation) and over time following LPS stimulation, where it engages these proresolving programs.42–45 We therefore investigated whether mitochondrial fragmentation and fusion regulate macrophage anti-inflammatory/proresolving responses by altering Arg1 expression following both pro- and anti-inflammatory stimuli. In unstimulated macrophages, preventing mitochondrial fusion increased Arg1 gene expression which was exacerbated upon LPS stimulation. Arg1 was similarly increased with IL-4 stimulation. Macrophage phagocytic capacity, a key function of proresolving macrophages, was also elevated when mitochondrial fusion was impaired. Our data demonstrate that, by promoting a proresolving macrophage phenotype with IL-4 or using a phenocopy siRNA approach, a fragmented mitochondrial network promotes Arg1 expression and proresolving functions in macrophages.

Inflammatory stimulation in macrophages results in high rates of glycolysis as a primary form of metabolism. This results in the accumulation of lactate through lactate dehydrogenase conversion of pyruvate. Recently, lactate and its derivative lactyl CoA have been attributed to a novel form of histone modification known as lactylation, which activates homeostatic reparatory pathways producing proresolving stimuli such as Arg1.31,37–39,46 Given that promoting mitochondrial fragmentation increased Arg1 expression following inflammatory stimulation, we hypothesized that this could be a result of increased lactate accumulation and histone lactylation. Indeed, knocking down the mitochondrial fusion machinery resulted in increased lactate and KLA levels after of LPS stimulation. The observed increase in lactate could be a result of decreased pyruvate conversion to acetyl co-A through the pyruvate (PDH) pathway, resulting in more pyruvate conversion to lactate via the lactate dehydrogenase (LDH) pathway. Indeed, inhibition of mitochondrial fusion resulted in decreased PDH protein expression, and subsequent blocking of LDH activity prevented the increase in Arg1 expression. Recently, PDH and its inhibitor PDHK have been associated with mitochondrial morphology in the setting of NLRP3 inflammasome activation, which aligns with our findings.36,40,47 In summary, by promoting mitochondrial fission in a pro-inflammatory setting results in a shift towards the M2 state through inhibition of PDH, therefore, increasing lactate levels, allowing for histone lactylation and the induction of proresolving factors such as Arg1.

Recently, LPS was shown to induce a fragmented mitochondrial phenotype, which correlates with the increased glycolysis associated with M1 polarization.16,17 In contrast, in the present study, we found M1 macrophages have a distinctly elongated mitochondrial network. We evaluated mitochondrial length by staining for an outer membrane protein, Tom20, which is (i) not subject to changes in mitochondrial leakage (as is the case with Mitotracker), (ii) distinct from other measures of inner mitochondrial membrane proteins that may change with metabolic shifts (i.e. complex I) and (iii) more robustly reflects the mitochondrial membrane structure and length.13,48,49 In addition, mitochondrial dyes used in live cell imaging can be toxic to cells and thus cause additional mitochondrial stress, which may be exacerbated upon co-stimulation with inflammatory factors like LPS. We performed additional analyses of mitochondrial length and dynamism using transmission electron microscopy (TEM) where we quantified cristae width and the ratio of mitochondria undergoing dynamic events vs static mitochondria, both of which increased in M1-like macrophages. Elongated mitochondrial networks were also seen in M1 polarized human THP1 macrophages, in murine BMDMs upon pro-inflammatory oxidized low-density lipoprotein (OxLDL) stimulation and in ex vivo peritoneal macrophages from mice fed a high-cholesterol diet for 2 weeks (data not shown). In addition, using all the same characterization methods, we found that M2 macrophages have a fragmented mitochondrial morphology. In conclusion, while specific methodologies used across different studies likely account for the different findings of mitochondrial morphology, we conclude that pro-inflammatory macrophages have a predominantly elongated mitochondrial network, whereas resting and M2 macrophages have a predominantly fragmented mitochondrial network.

Previous studies have demonstrated a role for key mitochondrial fission and fusion proteins, namely Drp1, Mfn2 and Opa1, in various aspects of macrophage inflammatory responses, sometimes with disparate results. Genetic deletion of Drp1- a critical mediator of mitochondrial fission- leads to reduced inflammatory responses measured by cytokine expression.16 Similarly, genetic deletion of either Mfn2 or Opa1- each of whom promote mitochondrial fusion also results in reduced inflammatory gene activation. Collectively, these data suggest that macrophages require a functional and dynamic mitochondrial network to maintain inflammatory responses. It is possible that deletion of a single member of the fission or fusion machinery may have nonmitochondrial effects (e.g. the nonmitochondrial functions of Drp1 are also lost) and/or complete loss of these key proteins may result in adaptations that alter cell function. Using an alternative approach, where siRNA knockdown of all three major fission or fusion machinery, we similarly observed that loss of functional mitochondrial fission or fusion resulted in impaired cytokine expression in unstimulated cells. However, we observed that loss of mitochondrial fusion in response to LPS resulted in increased Il1b gene expression and secretion, despite a decrease in the canonical NFκB subunit, p65. Indeed, genetic deletion of Opa1, which also demonstrated reduced p65, also resulted in increased IL-1β secretion.15 The authors did not discuss these discrepant findings, but our studies reveal that ATF4 and phospho-c-Jun alternative pro-inflammatory transcription factors are increased when mitochondrial fusion is lost. Therefore, despite lower baseline levels of cytokine expression, the lack of mitochondrial ability to undergo fusion activates the ATF4/c-Jun axis in response to LPS stimulation, which is likely an explanation for the elevated IL-1β in these macrophages. It is possible that loss of Opa1 throughout macrophage differentiation (i.e. genetic loss in bone-marrow-derived macrophage in culture) could impact inflammatory transcriptional pathway differently than the more acute loss of all three major fusion mediators, Opa1, Mfn1 and Mfn2, in mature macrophages. Nevertheless, these data collectively indicate that mitochondrial dynamics, particularly mitochondrial fusion, controls macrophage inflammatory responses.

Our model reveals that macrophages naturally adopt a fragmented mitochondrial phenotype over time following LPS stimulation, which corresponds to the activation of epigenetic changes and increase in arginase 1 expression and the adoption of a proresolving response. Moreover, this process can be accelerated by forcing fragmented mitochondrial phenotype. In the context of cancer, a more anti-inflammatory phenotype could lead to increased cancer growth/metastasis through impaired immune detection and a dampening of the cytotoxic inflammatory response. Indeed, tumor-associated macrophages often display M2-like characteristics and metabolism.50–53 In contrast, in the context of chronic inflammation like that which is found in atherosclerosis and obesity, blocking mitochondrial fusion or promoting mitochondrial fragmentation could promote tissue repair and more rapid resolution. Indeed, M1-like macrophages are abundant within the atherosclerotic plaque and obese adipose tissue, and previous reports have shown that Drp1-mediated mitochondrial fission is essential for efficient efferocytosis and atherosclerosis regression.54,55 We also see an elongated M1-like mitochondrial phenotype under lipid loading conditions both in vitro and ex vivo, suggesting that preventing mitochondrial fusion in these settings may be beneficial. This theory is supported by recent findings that hearts deficient in fusion machinery Mfn1 and Mfn2 are protected against acute myocardial infarction,56 and Opa1 deletion has been shown to increase macrophage persistence in muscle regeneration models.15 More work remains to be done to determine the disease and cell-specific context where impaired fusion could be of maximum benefit.

In conclusion, mitochondrial dynamics play a critical role in directing macrophage function and phenotype. Mitochondrial fusion uniquely controls the late-stage resolving phase of inflammation by controlling metabolite levels and proreparative gene expression programs.

MATERIALS AND METHODS

Animal procedures

C57BL/6 (wild-type, Charles River Laboratories) mice were maintained and housed in the University of Ottawa Heart Institute (UOHI) Animal Care and Veterinary facility. Mice ranged in age between 4 and 10 months with equal distribution of males and females.

Cell culture

Murine bone marrow-derived macrophages (BMDM) were obtained by flushing the femur and tibia in DMEM high glucose media (Gibco, 11965092) supplemented with 20% L929 conditioned media (a source of MCSF produced in-house from L929 cell culture), 10% heat-inactivated FBS (Gibco, 12483020) and 1% Antibiotic Antimycotic Solution (Gibco, 15240062). Monocytes were left to differentiate for 6 days in an incubator at 37 °C with 5% CO2. On day 6 the differentiated macrophages were lifted using 50 mM EDTA, counted and plated according to experimental conditions. All experimental procedures were started on day 7. Human monocytic THP-1 cell line were maintained in culture in RPMI medium (Gibco, 21870076) supplemented with 10% of heat-inactivated FBS (Gibco, 12483020), 10 mM Hepes (Gibco, 15630080), 10 mM L-Glutamine (Gibco, 25030149), 1 mM sodium pyruvate (Gibco, 11360070), 2.5 g/l D-glucose (Merck), 1% Penicillin-Streptomycin (Gibco, 15140122) and 50 pM β-mercaptoethanol (Gibco; 31350–010). THP-1 monocytes were differentiated into macrophages through incubation with 100 nM PMA (Sigma, P1585) in full media for 72 h.

Macrophage treatments

BMDM macrophages were polarized to M1 or M2 through treatment with 100 ng/mL LPS (Sigma, L4391) and 100 ng/mL IFN-γ (R&D, 485-MI-100) or 10 ng/mL IL-4 (R&D, 404-ML-010) for 24 h. THP-1 macrophages were polarized to M1 or M2 by incubation with 20 ng/mL IFN-γ (PeproTech, 300-02) and 100 ng/mL LPS (Sigma, L4391) or 20 ng/mL IL-4 (PeproTech, #200-04) and 20 ng/mL IL-13 (PeproTech, 200-13) for 24 h. LPS time courses were performed in full media with 100 ng/mL LPS (Sigma, L4391) with or without 10 mM sodium oxamate (SelleckChem, S6871). IL-4 time courses were performed in full media with 10 ng/mL IL-4 (R&D, 404-ML-010).

Knockdown

Fission knockdown macrophages were obtained by 72 h siRNA cotransfection with 40 nM Drp1 siRNA (Qiagen, SI0098226), 10 nM Mff siRNA (Qiagen, SI00855918) and 10 nM Fis1 siRNA (Qiagen, SI0145779) in OPTIMEM media (Gibco, 11058021) with Lipofectamine RNA iMAX transfection reagent (Life Technologies, 13778150) used according to manufacturers’ instructions. Fusion knockdown cells were obtained using the same method with 20 nM Opa1 siRNA (Qiagen, SI01365707), 20 nm Mfn1 siRNA (Qiagen, SI01304387), 20 nM Mfn2 siRNA (Qiagen, SI04392010). Knockdown efficiency was determined in comparison to control cells transfected with 60 nM negative control siRNA (Qiagen, 1027310).

RNA isolation & RT-qPCR

Total RNA was isolated using TRIzol (Invitrogen, 15596018) according to manufacturers instruction. The cDNA reactions were prepared from 750 ng of total RNA using iScript reverse transcription super mix (Bio-Rad, 1708841) and a 1:5 dilution was used for qPCR. Real-time quantitative PCR reactions were carried out using primer sequences obtained from the Harvard Medical School PrimerBank database (Table of primer sequences in Supplemental table 1) and SYBR green dye (Bio-Rad, 1725275). Data were normalized to either SRP14, TFAM or HPRT gene expression using the comparative ΔΔCt method and calculated fold changes relative to the controls.

Western blots

Total protein was extracted on ice in radioimmunoprecipitation assay buffer (RIPA) containing protease (Roche, 4693132001) and phosphatase (Roche, 4906837001) inhibitors. Proteins were separated on acrylamide gels of varying percentages and transferred to 0.22 μM polyvinylidene difluoride (PVDF) membranes. Membranes were blocked in 5% skim milk or 5% bovine serum albumin (BSA), depending on the antibody manufacturers’ specifications. Primary antibodies were incubated on the membrane overnight at 4 °C in 1% blocking. Membranes were incubated with LI-COR Secondaries (IRDye 800CW α-Rabbit and IRDye 680RD α-Mouse) at room temperature for an hour and imaged on the LI-COR Odyssey imaging system. Protein bands were quantified on ImageJ and normalized to housekeeping bands, either HSP90 or β-Actin. Proteins ATF4, Phos C-Jun, KLA and ARG1 required the use of HRP secondaries and chemiluminescence to be detected. Full list of antibodies and their catalogue numbers can be found in Supplemental Table 2.

Immunofluorescence microscopy

For all immunofluorescence microscopy, the macrophages were plated on glass coverslips within a 24-well plate. At the end of treatment (either polarization or siRNA transfection), the macrophages were fixed with 4% paraformaldehyde for 15 min at 37 °C, followed by 10 min of quenching with 10 mM NH4Cl. The cells were permeabilized with 0.1% Triton X-100 for 10 min. 10% FBS was used as a blocking agent and incubated on the coverslips for 30 min. Primary antibodies were then incubated for an hour in 5% FBS followed by secondary antibodies also in 5% FBS for an hour. Primary antibodies used are detailed in the associated figure legends. Secondaries used were Alexa Fluor 555 or 488 in the associated species. 1 μg/mL DAPI (Becton Dickinson, 564907) was used as a nuclear dye and incubated on the coverslips for 5 min. Coverslips were then washed and mounted using Dako Fluorescence mounting media (Cedarlane, S3022380-2) on microscopy slides (Fisher Scientific, 12-552). Cells were imaged within 2 days at 63× on a Zeiss LSM 880 microscope. Mitochondrial length was measured manually using the ImageJ software and presented as mean length (μM) per cell. HIF-1α nuclear translocation was also measured on ImageJ, whereby we quantified the total mean fluorescence intensity (MFI) per cell and the MFI within the nucleus. Data is presented as a ratio of nuclear/cytoplasmic MFI per cell.

Transmission electron microscopy

Cells were fixed for 60 min in 2% PFA and 2.5% glutaraldehyde. The samples were processed and imaged at the University of Ottawa Heart Institute Cell Imaging and Histology Core Facility according to standard protocols. Those images were quantified for mitochondrial length and cristae width using the ImageJ software. All dynamic events (either fission or fusion) were quantified as a ratio between static vs dynamic mitochondria per cell.

Phagocytosis assay

Cells plated on glass coverslips within a 24-well plate were incubated with equal concentrations of polystyrene fluoresbrite YG microspheres (Polysciences, 17154-10) for 2 h at 37 °C. The wells were washed with PBS to remove excess beads, then fixed and stained for Tom20 according to the above IF protocol. The cells were imaged within 2 days at 63× on a Zeiss LSM 880 microscope. Images were manually quantified for the number of internalized fluorescent beads per cell on ImageJ.

Il-1β ELISA

Fission and fusion knockdown cells were assessed for changes in IL-1β secretion upon LPS (100 ng/μL) stimulation with and without 5 mM ATP (Bio-Rad, 1725275). Cell media was collected at end of LPS treatment centrifuged at 1200 g for 5 min to discard any cell debris. Media was quantified for secreted IL-1β by mouse IL-1 beta Quantikine ELISA Kit (R&D, MLB00C) according to the manufacturer’s instruction.

Lactate levels

Fusion knockdown cell lysate lactate levels were quantified using the L-Lactate Assay Kit (ab65331) according to manufacturers’ instructions.

Statistical analysis

All data involving statistics are presented as mean ± SEM. The same control was used for individual comparisons to fission KD or fusion KD. The statistical significance of the differences between groups was determined on Prism V9.5.0 software (GraphPad Software Inc.) using unpaired Student’s t tests, multiple unpaired t tests or one-way ANOVA. Normal distribution was determined by the Shapiro–Wilk test. The number of replicates and the statistical test used are described in the figure legends.

Supplementary Material

Supplemental Material

Funding Statement

This work was supported by grant funding to KJR from the Canadian Institutes of Health Research and the Heart & Stroke Foundation of Canada.

DATA AVAILABILITY STATEMENT

The data that support the findings of this study are available here: https://figshare.com/projects/Susser_et_al_Mitochondrial_fragmentation_promotes_inflammation_resolution_responses_in_macrophages_via_histone_lactylation_/176217

DISCLOSURE STATEMENT

No potential conflict of interest was reported by the author(s).

REFERENCES

  • 1.Moore KJ, Tabas I.. Macrophages in the pathogenesis of atherosclerosis. Cell. 2011;145:341–355. PMID: 21529710 doi: 10.1016/j.cell.2011.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Rodríguez-Prados J-C, Través PG, Cuenca J, Rico D, Aragonés J, Martín-Sanz P, Cascante M, Boscá L.. Substrate fate in activated macrophages: a comparison between innate, classic, and alternative activation. J Immunol. 2010;185:605–614. PMID: 20498354 doi: 10.4049/jimmunol.0901698. [DOI] [PubMed] [Google Scholar]
  • 3.Murray PJ. Macrophage polarization. Annu Rev Physiol. 2017;79:541–566. PMID: 27813830 doi: 10.1146/annurev-physiol-022516-034339. [DOI] [PubMed] [Google Scholar]
  • 4.Tabas I, Bornfeldt KE.. Macrophage phenotype and function in different stages of atherosclerosis. Circ Res. 2016;118:653–667. PMID: 26892964 doi: 10.1161/CIRCRESAHA.115.306256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Libby P. Vascular biology of atherosclerosis: overview and state of the art. Am J Cardiol. 2003;91:3A–6A. doi: 10.1016/S0002-9149(02)03143-0.PMID: 12645637 [DOI] [PubMed] [Google Scholar]
  • 6.Moore KJ, Sheedy FJ, Fisher EA.. 2013. Macrophages in atherosclerosis: a dynamic balance. Nat Rev Immunol. 2013;13:709–721. PMID: 23995626 doi: 10.1038/nri3520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Ravi S, Mitchell T, Kramer PA, Chacko B, Darley-Usmar VM.. Mitochondria in monocytes and macrophages-implications for translational and basic research. Int J Biochem Cell Biol. 2014;53:202–207. PMID: 24863362 doi: 10.1016/j.biocel.2014.05.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Wculek SK, Dunphy G, Heras-Murillo I, Mastrangelo A, Sancho D.. Metabolism of tissue macrophages in homeostasis and pathology. Cell Mol Immunol. 2022;19:384–408. PMID: 34876704 doi: 10.1038/s41423-021-00791-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Friedman JR, Nunnari J.. Mitochondrial form and function. Nature. 2014;505:335–343. PMID: 24429632 doi: 10.1038/nature12985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Polyakov VY, Soukhomlinova MY, Fais D.. Fusion, fragmentation, and fission of mitochondria. Biochemistry (Mosc). 2003;68:838–849. PMID: 12948383 doi: 10.1023/a:1025738712958. [DOI] [PubMed] [Google Scholar]
  • 11.Suárez-Rivero JM, Villanueva-Paz M, Cruz-Ojeda P d l, Mata M d l, Cotán D, Oropesa-Ávila M, Lavera I d, Álvarez-Córdoba M, Luzón-Hidalgo R, Sánchez-Alcázar JA.. Mitochondrial dynamics in mitochondrial diseases. Diseases. 2016;5:1. doi: 10.3390/diseases5010001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Archer SL. Mitochondrial dynamics - mitochondrial fission and fusion in human diseases. N Engl J Med. 2013;369:2236–2251. PMID: 24304053 doi: 10.1056/NEJMra1215233. [DOI] [PubMed] [Google Scholar]
  • 13.Khacho M, Clark A, Svoboda DS, Azzi J, MacLaurin JG, Meghaizel C, Sesaki H, Lagace DC, Germain M, Harper M-E, et al. . Mitochondrial dynamics impacts stem cell identity and fate decisions by regulating a nuclear transcriptional program. Cell Stem Cell. 2016;19:232–247. PMID: 27237737 doi: 10.1016/j.stem.2016.04.015. [DOI] [PubMed] [Google Scholar]
  • 14.Baker N, Wade S, Triolo M, Girgis J, Chwastek D, Larrigan S, Feige P, Fujita R, Crist C, Rudnicki MA, et al. . The mitochondrial protein OPA1 regulates the quiescent state of adult muscle stem cells. Cell Stem Cell. 2022;29:1315–1332.e9. PMID: 35998642 doi: 10.1016/j.stem.2022.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Sánchez-Rodríguez R, Tezze C, Agnellini AHR, Angioni R, Venegas FC, Cioccarelli C, Munari F, Bertoldi N, Canton M, Desbats MA, et al. . 2023. OPA1 drives macrophage metabolism and functional commitment via p65 signaling. Cell Death Differ. 2023;30:742–752. PMID: 36307526 doi: 10.1038/s41418-022-01076-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Kapetanovic R, Afroz SF, Ramnath D, Lawrence GM, Okada T, Curson JE, Bruin J, Fairlie DP, Schroder K, St John JC, et al. . Lipopolysaccharide promotes Drp1‐dependent mitochondrial fission and associated inflammatory responses in macrophages. Immunol Cell Biol. 2020;98:528–539. PMID: 32686869 doi: 10.1111/imcb.12363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Gao F, Reynolds MB, Passalacqua KD, Sexton JZ, Abuaita BH, O'Riordan MXD.. 2021. The mitochondrial fission regulator DRP1 controls post-transcriptional regulation of TNF-α. Front Cell Infect Microbiol. 2020;10:593805. doi: 10.3389/fcimb.2020.593805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Bordt EA, Clerc P, Roelofs BA, Saladino AJ, Tretter L, Adam-Vizi V, Cherok E, Khalil A, Yadava N, Ge SX, et al. . The putative Drp1 inhibitor mdivi-1 Is a Reversible Mitochondrial Complex I inhibitor that modulates reactive oxygen species. Dev Cell. 2017;40:583–594.e6. PMID: 28350990 doi: 10.1016/j.devcel.2017.02.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Buck MD, O'Sullivan D, Klein Geltink RI, Curtis JD, Chang C-H, Sanin DE, Qiu J, Kretz O, Braas D, van der Windt GJW, et al. . 2016. Mitochondrial dynamics controls T cell fate through metabolic programming. Cell. 2016;166:63–76. PMID: 27293185 doi: 10.1016/j.cell.2016.05.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Kelley N, Jeltema D, Duan Y, He Y.. The NLRP3 inflammasome: an overview of mechanisms of activation and regulation. Int J Mol Sci. 2019;20:3328. PMID: 31284572 doi: 10.3390/ijms20133328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.He Y, Hara H, Núñez G.. Mechanism and regulation of NLRP3 inflammasome activation. Trends Biochem Sci. 2016;41:1012–1021. PMID: 27669650 doi: 10.1016/j.tibs.2016.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Ichinohe T, Yamazaki T, Koshiba T, Yanagi Y.. Mitochondrial protein mitofusin 2 is required for NLRP3 inflammasome activation after RNA virus infection. Proc Natl Acad Sci U S A. 2013;110:17963–17968. PMID: 24127597 doi: 10.1073/pnas.1312571110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Park S, Won J-H, Hwang I, Hong S, Lee HK, Yu J-W.. Defective mitochondrial fission augments NLRP3 inflammasome activation. Sci Rep. 2015;5:15489. PMID: 26489382 doi: 10.1038/srep15489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Dorrington MG, Fraser IDC.. NF-κB Signaling in macrophages: dynamics, crosstalk, and signal integration. Front Immunol. 2019;10:705. PMID: 31024544 doi: 10.3389/fimmu.2019.00705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Zhou R, Yazdi AS, Menu P, Tschopp J.. A role for mitochondria in NLRP3 inflammasome activation. Nature. 2011;469:221–225. PMID: 21124315 doi: 10.1038/nature09663. [DOI] [PubMed] [Google Scholar]
  • 26.Lu Y-C, Yeh W-C, Ohashi PS.. LPS/TLR4 signal transduction pathway. Cytokine. 2008;42:145–151. PMID: 18304834 doi: 10.1016/j.cyto.2008.01.006. [DOI] [PubMed] [Google Scholar]
  • 27.Corcoran SE, O'Neill LAJ.. HIF1α and metabolic reprogramming in inflammation. J Clin Invest. 2016;126:3699–3707. PMID: 27571407 doi: 10.1172/JCI84431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Zhang C, Bai N, Chang A, Zhang Z, Yin J, Shen W, Tian Y, Xiang R, Liu C.. ATF4 is directly recruited by TLR4 signaling and positively regulates TLR4-trigged cytokine production in human monocytes. Cell Mol Immunol. 2013;10:84–94. PMID: 23241898 doi: 10.1038/cmi.2012.57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Koppula P, Zhuang L, Gan B.. Cystine transporter SLC7A11/xCT in cancer: ferroptosis, nutrient dependency, and cancer therapy. Protein Cell. 2021;12:599–620. PMID: 33000412 doi: 10.1007/s13238-020-00789-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Pourcet B, Pineda-Torra I.. Transcriptional regulation of macrophage arginase 1 expression and its role in atherosclerosis. Trends Cardiovasc Med. 2013;23:143–152. PMID: 23375628 doi: 10.1016/j.tcm.2012.10.003. [DOI] [PubMed] [Google Scholar]
  • 31.Dichtl S, Lindenthal L, Zeitler L, Behnke K, Schlösser D, Strobl B, Scheller J, El Kasmi KC, Murray PJ.. Lactate and IL6 define separable paths of inflammatory metabolic adaptation. Sci Adv. 2021;7:3505–3528. PMID: 34162546 doi: 10.1126/sciadv.abg3505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Bhattacharjee A, Shukla M, Yakubenko VP, Mulya A, Kundu S, Cathcart MK.. IL-4 and IL-13 employ discrete signaling pathways for target gene expression in alternatively activated monocytes/macrophages. Free Radic Biol Med. 2013;54:1–16. doi: 10.1016/j.freeradbiomed.2012.10.553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Yunna C, Mengru H, Lei W, Weidong C.. Macrophage M1/M2 polarization. Eur J Pharmacol. 2020;877:173090. PMID: 32234529 doi: 10.1016/j.ejphar.2020.173090. [DOI] [PubMed] [Google Scholar]
  • 34.Liu Y, Xu R, Gu H, Zhang E, Qu J, Cao W, Huang X, Yan H, He J, Cai Z.. Metabolic reprogramming in macrophage responses. Biomark Res. 2021;9:1. PMID: 33407885 doi: 10.1186/s40364-020-00251-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Li X, Yang Y, Zhang B, Lin X, Fu X, An Y, Zou Y, Wang J-X, Wang Z, Yu T.. Lactate metabolism in human health and disease. Signal Transduct Target Ther. 2022;7:305. PMID: 36050306 doi: 10.1038/s41392-022-01151-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Noe JT, Rendon BE, Geller AE, Conroy LR, Morrissey SM, Young LEA, Bruntz RC, Kim EJ, Wise-Mitchell A, Barbosa de Souza Rizzo M, et al. . Lactate supports a metabolic-epigenetic link in macrophage polarization. Sci Adv. 2021;7:eabi8602. doi: 10.1126/sciadv.abi8602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Chen A-N, Luo Y, Yang Y-H, Fu J-T, Geng X-M, Shi J-P, Yang J.. Lactylation, a novel metabolic reprogramming code: current status and prospects. Front Immunol. 2021;12:688910. PMID: 34177945 doi: 10.3389/fimmu.2021.688910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Xin Q, Wang H, Li Q, Liu S, Qu K, Liu C, Zhang J.. Lactylation: a passing fad or the future of posttranslational modification. Inflammation. 2022;45:1419–1429. PMID: 35224683 doi: 10.1007/s10753-022-01637-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Zhang D, Tang Z, Huang H, Zhou G, Cui C, Weng Y, Liu W, Kim S, Lee S, Perez-Neut M, et al. . Metabolic regulation of gene expression by histone lactylation. Nature. 2019;574:575–580. PMID: 31645732 doi: 10.1038/s41586-019-1678-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Meyers AK, Wang Z, Han W, Zhao Q, Zabalawi M, Duan L, Liu J, Zhang Q, Manne RK, Lorenzo F, et al. . Pyruvate dehydrogenase kinase supports macrophage NLRP3 inflammasome activation during acute inflammation. Cell Rep. 2023;42:111941. PMID: 36640341 doi: 10.1016/j.celrep.2022.111941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Khacho M, Harris R, Slack RS.. Mitochondria as central regulators of neural stem cell fate and cognitive function. Nat Rev Neurosci. 2019;20:34–48. PMID: 30464208 doi: 10.1038/s41583-018-0091-3. [DOI] [PubMed] [Google Scholar]
  • 42.Rath M, Müller I, Kropf P, Closs EI, Munder M.. Metabolism via Arginase or Nitric Oxide Synthase: Two Competing Arginine Pathways in Macrophages. Front Immunol. 2014;5:532. doi: 10.3389/fimmu.2014.00532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Yang Z, Ming X-F.. Functions of arginase isoforms in macrophage inflammatory responses: impact on cardiovascular diseases and metabolic disorders. Front Immunol. 2014;5:533. PMID: 25386179 doi: 10.3389/fimmu.2014.00533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Wang X, Chen Y, Qin W, Zhang W, Wei S, Wang J, Liu FQ, Gong L, An FS, Zhang Y, et al. . Arginase I attenuates inflammatory cytokine secretion induced by lipopolysaccharide in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol. 2011;31:1853–1860. PMID: 21617140 doi: 10.1161/ATVBAHA.111.229302. [DOI] [PubMed] [Google Scholar]
  • 45.El Kasmi KC, Qualls JE, Pesce JT, Smith AM, Thompson RW, Henao-Tamayo M, Basaraba RJ, König T, Schleicher U, Koo M-S, et al. . Toll-like receptor–induced arginase 1 in macrophages thwarts effective immunity against intracellular pathogens. Nat Immunol. 2008;9:1399–1406. PMID: 18978793 doi: 10.1038/ni.1671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Manoharan I, Prasad PD, Thangaraju M, Manicassamy S.. Lactate-dependent regulation of immune responses by dendritic cells and macrophages. Front Immunol. 2021;12:691134. PMID: 34394085 doi: 10.3389/fimmu.2021.691134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Thoudam T, Chanda D, Sinam IS, Kim BG, Kim MJ, Oh CJ, Lee JY, Kim MJ, Park SY, Lee SY, et al. . Noncanonical PDK4 action alters mitochondrial dynamics to affect the cellular respiratory status. Proc Natl Acad Sci U S A. 2022;119:e2120157119. PMID: 35969774 doi: 10.1073/PNAS.2120157119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Patten DA, Wong J, Khacho M, Soubannier V, Mailloux RJ, Pilon‐Larose K, MacLaurin JG, Park DS, McBride HM, Trinkle‐Mulcahy L, et al. . OPA1‐dependent cristae modulation is essential for cellular adaptation to metabolic demand. Embo J. 2014;33:2676–2691. PMID: 25298396 doi: 10.15252/embj.201488349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Khacho M, Tarabay M, Patten D, Khacho P, MacLaurin JG, Guadagno J, Bergeron R, Cregan SP, Harper M-E, Park DS, et al. . Acidosis overrides oxygen deprivation to maintain mitochondrial function and cell survival. Nat Commun. 2014;5:3550. PMID: 24686499 doi: 10.1038/ncomms4550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Pan Y, Yu Y, Wang X, Zhang T.. Tumor-Associated Macrophages in Tumor Immunity. Front Immunol. 2020;11:583084. PMID: 33365025 doi: 10.3389/fimmu.2020.583084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Vitale I, Manic G, Coussens LM, Kroemer G, Galluzzi L.. Macrophages and metabolism in the tumor microenvironment. Cell Metab. 2019;30:36–50. PMID: 31269428 doi: 10.1016/j.cmet.2019.06.001. [DOI] [PubMed] [Google Scholar]
  • 52.Li J, DeNicola GM, Ruffell B.. Metabolism in tumor-associated macrophages. Int Rev Cell Mol Biol. 2022;367;65–100. doi: 10.1016/bs.ircmb.2022.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Zhang Q, Wang J, Yadav DK, Bai X, Liang T.. Glucose metabolism: the metabolic signature of tumor associated macrophage. Front Immunol. 2021;12:702580. PMID: 34267763 doi: 10.3389/fimmu.2021.702580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Yurdagul A, Subramanian M, Wang X, Crown SB, Ilkayeva OR, Darville L, Kolluru GK, Rymond CC, Gerlach BD, Zheng Z, et al. . Macrophage metabolism of apoptotic cell-derived arginine promotes continual efferocytosis and resolution of injury. Cell Metab. 2020;31:518–533.e10. PMID: 32004476 doi: 10.1016/j.cmet.2020.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Wang Y, Subramanian M, Yurdagul A, Barbosa-Lorenzi VC, Cai B, de Juan-Sanz J, Ryan TA, Nomura M, Maxfield FR, Tabas I.. Mitochondrial fission promotes the continued clearance of apoptotic cells by macrophages. Cell. 2017;171:331–345.e22. PMID: 28942921 doi: 10.1016/j.cell.2017.08.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Hall AR, Burke N, Dongworth RK, Kalkhoran SB, Dyson A, Vicencio JM, Dorn GW, Yellon DM, Hausenloy DJ.. Hearts deficient in both Mfn1 and Mfn2 are protected against acute myocardial infarction. Cell Death Dis. 2016;7:e2238–e2238–e2238. PMID: 27228353 doi: 10.1038/cddis.2016.139. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplemental Material

Data Availability Statement

The data that support the findings of this study are available here: https://figshare.com/projects/Susser_et_al_Mitochondrial_fragmentation_promotes_inflammation_resolution_responses_in_macrophages_via_histone_lactylation_/176217


Articles from Molecular and Cellular Biology are provided here courtesy of Taylor & Francis

RESOURCES