Next Article in Journal
ZEB1 and Neural Stem Cells: Insights into Microglia-Conditioned Medium-Driven Neuroinflammation
Previous Article in Journal
Adenosine Receptors in Neuroinflammation and Neurodegeneration
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immunoproteasomes in Skeletal Muscle Pathologies: Emerging Roles, Conflicting Evidence, and Future Directions

by
Alexander Kalinkovich
1 and
Gregory Livshits
1,2,*
1
Department of Anatomy and Anthropology, Gray Faculty of Medical and Health Sciences, Tel-Aviv University, Tel Aviv 6927846, Israel
2
Department of Morphological Sciences, Adelson School of Medicine, Ariel University, Ariel 4077625, Israel
*
Author to whom correspondence should be addressed.
Cells 2025, 14(20), 1586; https://doi.org/10.3390/cells14201586
Submission received: 8 September 2025 / Revised: 6 October 2025 / Accepted: 11 October 2025 / Published: 12 October 2025

Abstract

Highlights

What are the main findings?
  • Immunoproteasomes (IMPs) act as a double-edged sword in muscle biology; they promote protein quality control and muscle differentiation under normal or early disease conditions, but they can drive muscle wasting when persistently activated.
  • Excessive IMP activity fosters chronic inflammation and harmful crosstalk between immune and muscle cells, thereby worsening degeneration.
What is the implication of the main findings?
  • Targeting IMPs with selective inhibitors holds therapeutic promise in skeletal muscle diseases and experimental models of muscle loss.
  • However, in skeletal muscle disorders, inhibition may be a mixed blessing, potentially reducing damaging inflammation but at the cost of impairing muscle maintenance and repair.

Abstract

Skeletal muscle pathologies, including sarcopenia, inflammatory myopathies, and various muscular dystrophies, are strongly influenced by chronic low-grade inflammation and impaired proteostasis. Immunoproteasomes (IMPs), inducible proteolytic complexes activated by pro-inflammatory cytokines, are emerging as regulators linking immune signaling to protein quality control. Evidence suggests that IMPs have paradoxical, context-dependent roles in skeletal muscle. On one hand, they can support proteostasis and muscle regeneration under stress; on the other, persistent activation may sustain cytokine production, antigen presentation, and maladaptive immune–muscle interactions, promoting chronic inflammation and muscle wasting. Selective IMP inhibitors, such as ONX 0914 and KZR-616, display potent anti-inflammatory effects in preclinical models of autoimmune myositis and muscle atrophy. Yet, their use in skeletal muscle pathologies is controversial; while inhibition may dampen harmful immune activation, it could also impair muscle repair and proteostasis. This review summarizes current findings, highlights key contradictions, and explores unresolved questions about the role of IMPs in skeletal muscle pathologies. We emphasize the need for a deeper understanding of IMP-mediated mechanisms in skeletal muscle pathology and strategies combining selective inhibitors to enhance therapeutic efficacy while minimizing adverse effects. IMPs thus represent both a promising and potentially risky therapeutic target, with outcomes highly dependent on disease context.

1. Introduction

Sarcopenia is an age-related condition characterized by the progressive loss of skeletal muscle mass, strength, and function, and it is associated with serious adverse health outcomes, including increased risk of falls and fractures, functional decline, elevated healthcare costs, and mortality [1,2,3]. Although the etiology of sarcopenia is multifactorial [4], chronic low-grade inflammation has emerged as a central factor in its pathogenesis. This persistent inflammatory state contributes to muscle catabolism, oxidative stress, and impaired regenerative capacity not only in sarcopenia [5,6,7,8,9] but also in various muscular dystrophies [10] and inflammatory myopathies, such as polymyositis and dermatomyositis [11].
Imbalanced muscle protein turnover is another hallmark of sarcopenia [12] and other skeletal muscle pathologies [13,14]. This dysregulation is primarily governed by the ubiquitin–proteasome system (UPS), a highly conserved proteolytic pathway responsible for degrading damaged, misfolded, or short-lived regulatory proteins. Within the UPS, polyubiquitinated proteins are recognized and degraded by the 26S proteasome into short peptides and amino acids, thereby maintaining cellular proteostasis and preventing proteotoxic stress [15,16,17,18].
A specialized form of the proteasome, the immunoproteasome (IMP), is primarily expressed in immune cells, such as dendritic cells, macrophages, T lymphocytes, and natural killer cells, but it can also be induced in non-immune cells under inflammatory conditions. In IMPs, the constitutive catalytic β subunits β1, β2, and β5 are replaced by the inducible subunits β1i (LMP2), β2i (MECL1), and β5i (LMP7), respectively. Assembly of IMP is triggered by pro-inflammatory cytokines, particularly interferon-γ (IFNγ) and tumor necrosis factor-α (TNFα), which promote the substitution of the standard catalytic subunits with their inducible counterparts. This modification enhances the degradation of oxidized proteins and improves the generation of peptides for presentation on major histocompatibility complex class I (MHC-I) molecules, thus contributing to immune surveillance [19,20,21]. Beyond their role in immunosurveillance, accumulating evidence implicates IMPs in broader immunomodulatory functions, including the regulation of cytokine production, macrophage polarization, NLRP3 inflammasome activity, and inflammatory tissue damage [22,23].
However, within skeletal muscle biology, IMPs appear to play a nuanced and context-dependent role. Evidence suggests that skeletal muscle does express IMPs; for example, muscle biopsies from older adults have shown upregulation of IMP subunits, particularly β5i (LMP7), which correlates with increased expression of pro-inflammatory cytokines, such as IL-6 and TNFα [24]. Functional studies further support a role for IMPs in muscle homeostasis. Specifically, knockdown or pharmacological inhibition of β1i (LMP2) impairs C2C12 myoblast differentiation, increases protein oxidation and apoptosis, and inhibits myotube formation, highlighting a critical role for IMPs in myogenesis [25]. In vivo, β5i (LMP7) has been shown to support skeletal muscle regeneration by promoting the macrophage phenotype switch from pro-inflammatory (M1) to anti-inflammatory (M2). Inhibition of β5i (LMP7) in mice impairs muscle regeneration and increases interstitial fibrosis, further suggesting a beneficial role of IMPs in muscle repair [26].
Conversely, in certain pathological contexts, IMP activity appears detrimental to skeletal muscle function. In a murine model of muscular dystrophy, IMP inhibition alleviated muscle weakness and atrophy, likely through a reduction in muscle inflammation [27]. Similarly, in idiopathic inflammatory myopathies (IIMs), such as polymyositis and dermatomyositis, disorders characterized by immune-mediated muscle damage, akin in part to sarcopenia, there is constitutive overexpression of IMP subunits β1i (LMP2), β2i (MECL1), and β5i (LMP7) in affected muscle tissue [28,29]. Notably, in a mouse model of polymyositis, selective IMP inhibition preserved grip strength and reduced leukocyte infiltration into muscle, indicating a potentially harmful role for IMPs in inflammatory myopathies [30].
Despite these findings, no direct studies to date have examined the regulation of IMPs in sarcopenic muscle or their mechanistic connection to chronic inflammation. It remains unresolved whether IMP upregulation in aging muscle represents a compensatory mechanism aimed at maintaining proteostasis and supporting regeneration or a maladaptive response that exacerbates chronic inflammation, paralleling the pathological features observed in IIMs. An additional open question is whether IMP inhibition, while beneficial in inflammatory myopathies, would impair muscle regeneration in the context of sarcopenia due to the IMP’s role in myogenesis, or, conversely, whether it might attenuate chronic inflammation and thus help preserve muscle function in skeletal muscle pathologies.
This review aims to dissect the complex, context-dependent roles of immunoproteasomes in skeletal muscle pathologies, including but not limited to sarcopenia. We analyze current evidence on their involvement in regulating proteostasis, immune signaling, and inflammation and critically assess the therapeutic promise and risks of selective IMP inhibitors. By identifying key contradictions and unresolved questions, we seek to guide future strategies that balance immune modulation with the preservation of muscle function.

2. Immunoproteasomes: A Brief Overview

2.1. Structure

The structure of proteasomes and their role in proteostasis have been extensively described in numerous review articles, both under physiological conditions and in the context of various diseases, particularly cancer and neurodegenerative disorders (e.g., [31,32,33]). Much less is known about IMPs, although emerging evidence suggests their involvement not only in virus-induced inflammation [22,34] but also in disorders associated with chronic inflammation, such as autoimmune diseases [35,36,37] and neuroinflammatory disorders [38]. In this section, we briefly discuss the basic terms relevant to this article.
The proteasome is a critical component in the final step of protein degradation within cells, particularly within the UPS. It acts as a large, multi-subunit protease complex that degrades polyubiquitinated old, unused, and damaged proteins into peptides 8–21 amino acid long, which are further cleaved into amino acids for reuse by the ribosome [39]. As diagrammatically illustrated in Figure 1, the constitutive proteasome consists of a combination of two α and two β rings, each containing seven subunits. The β ring includes three standard catalytic subunits, β1c, β2c and β5c, each possessing caspase-like, trypsin-like, and chymotrypsin-like hydrolytic activity, respectively, which serve as the hydrolytic core of the proteasome [40,41]. In hematopoietic cells, including immune cells, and in cells stimulated with IFNγ, other pro-inflammatory molecules [42,43,44], or by oxidative stress [45], the catalytically active standard proteasome subunits β1c, β2c, and β5c in the inner two rings of the 20S proteasome are replaced by β1i (low molecular mass polypeptide 2 (LMP2, encoded by PSMB9), β2i (multicatalytic endopeptidase complex–like 1 (MECL-1), encoded by PSMB10), and β5i (LMP7, encoded by PSMB8), forming the IMP. In IMPs, proteasome activators PA700 (19S proteasome), PA28 α/β (11S proteasome), or a combination of both bind to 20S at its two ends. Notably, in IPMs, the caspase-like activity, exerted by β1c in standard proteasomes, is strongly reduced, whereas the chymotrypsin-like activity is enhanced [46,47,48]. This results in the generation of peptides with hydrophobic C-terminal residues, which are well-suited for presentation to MHC-I molecules [21].
As mentioned above, IMPs incorporate three inducible catalytic subunits, β1i (LMP2), β2i (MECL-1), and β5i (LMP7), which differ from their constitutive counterparts in both sequence (~60% identity) and substrate specificity, producing peptides better suited for MHC class I antigen presentation [49]. Importantly, IMPs do not form through subunit exchange in pre-existing 20S proteasomes. Instead, they assemble de novo following the induction of βi subunit expression. β1i is typically incorporated first, promoting the sequential addition of β2i and β5i [20,50]. This order contrasts with constitutive proteasome assembly, where β2 is usually incorporated first [51]. The selective incorporation of βi subunits is largely driven by their distinct propeptides, which influence chaperone interactions and subunit selection. Notably, IMP assembly is significantly faster, up to four times, than that of constitutive proteasomes, due in part to direct binding of β5i to the chaperone proteasome maturation protein (POMP) [38]. As a result of this coordinated assembly, most proteasomes in IFNγ-stimulated cells contain all three βi subunits, ensuring that IMPs dominate under inflammatory conditions [20,52,53].
Notably, there are currently no quantitative data available on the ratio between IMPs and constitutive 20S proteasomes in muscle fibers (or in other cell types) following exposure to pro-inflammatory stimuli or even under non-induced conditions. This gap in the literature makes it difficult to precisely assess how IMPs dynamically replace or coexist with standard proteasomes in these contexts [54]. This issue is further complicated by conflicting findings. Some studies suggest that IMPs more efficiently degrade polyubiquitinated proteins under IFNγ–induced inflammatory conditions [55], while others report no significant difference between standard and IMPs in this function [56]. These inconsistencies highlight the need for more precise, quantitative studies to clarify IMP dynamics and function in skeletal muscle.
The proteasomal core is composed of four stacked rings, two outer α rings and two inner β rings, each consisting of seven subunits. In response to pro-inflammatory stimuli, such as IFNγ, IL-1β, or TNFα, the β1, β2, and β5 catalytic subunits of the constitutive 20S proteasome are replaced by their immunoproteasome-specific counterparts, β1i, β2i, and β5i, forming the 20S immunoproteasome. This core particle can then associate at one or both ends with proteasome activators, such as PA700 (19S regulatory particle), PA28α/β (11S activator), or a combination of both, resulting in the formation of fully functional immunoproteasomes. A summary of the genes encoding these catalytic subunits and their associated enzymatic activities in both the constitutive and immunoproteasome forms is presented in the table at the bottom of the diagram.

2.2. Function

Recent findings indicate that proteasomes of most immune cell subsets mainly consist of IMP subunits [57]. Apart from MHC-I antigen processing, many processes have been attributed to the IMPs, including T cell expansion [47,58,59], the generation of Th17 cells while promoting regulatory T cell (Treg) development [60,61,62], macrophage polarization [63,64], protection from immunopathological damage in the brain [65,66], and transplant rejection [67]. IMPs are also considered to be involved in the development of a wide range of inflammation-associated diseases, including lung-related [68,69,70], neurodegenerative [71,72], cardiovascular (CVD) [73,74], and autoimmune diseases [35,75].

Inhibitors

The potential involvement of IMPs in the pathogenesis of various inflammation-associated disorders, as discussed above, positions IMPs as attractive therapeutic targets. To date, both nonselective and selective IMP inhibitors have been developed for potential clinical applications. Bortezomib (Velcade®), a nonselective proteasome inhibitor, has been widely used in the treatment of multiple myeloma [76]. However, due to its lack of specificity, bortezomib inhibits not only the proteasome but also several off-target proteins. This broad activity contributes to a high incidence of adverse effects, particularly peripheral neuropathy, which affects more than 30% of treated patients [77].
A major breakthrough in the field occurred in 2009 with the discovery of ONX 0914 (formerly PR-957), a selective inhibitor of the IMP catalytic subunit LMP7 (β5i) [78] whose selectivity can be explained by the difference between the S1 pockets of β5 and β5i [79]. The study, which reported the efficacy of ONX 0914 in mouse models of autoimmune diseases, represented a paradigm shift and caused widespread interest in the field. For the first time, it provided proof of concept that selective IMP inhibition could attenuate inflammation without broadly suppressing proteasomal function. While primarily targeting LMP7, ONX 0914 also inhibits LMP2 and MECL-1 subunits, though to a lesser extent [80]. Importantly, ONX 0914 exhibits at least 14-fold greater selectivity for LMP7 over β5c and does not significantly impair overall proteasome function.
In animal models, it effectively inhibited LMP7-dependent antigen presentation and suppressed the release of key pro-inflammatory cytokines, including IL-23, IFNγ, and IL-2, thereby demonstrating strong therapeutic potential [78]. Notably, ONX 0914 showed reduced systemic toxicity compared to nonselective proteasome inhibitors, underscoring its promise as a safer and more targeted therapeutic strategy for conditions associated with elevated IMP expression, such as chronic inflammatory diseases, neurodegenerative disorders, and certain cancers.
These seminal findings not only validated IMP as a druggable target but also sparked the development of additional IMP-specific compounds, such as KZR-616, a selective inhibitor of LMP2 and LMP7, which is 18- and 81-fold more selective for LMP7 and LMP2 than for their standard counterparts β5c and β1c, respectively, and is currently under clinical investigation for autoimmune and inflammatory conditions [81]. Another noteworthy inhibitor, M3258, is an orally bioavailable, potent, reversible, and highly selective LMP7 inhibitor with 305-fold greater selectivity for LMP7 over β5c. It has demonstrated a favorable safety profile in animal studies, with no observed adverse effects in vital organs, including the heart, lungs, kidneys, nervous system, and gastrointestinal tract [82]. In models of heart transplantation, DPLG3 has shown efficacy and demonstrated greater selectivity than ONX 0914 [83]. IPSI-001 has emerged as a more promising inhibitor that does not bind to β2c or β5c and exhibits a 100-fold preference for IMPs with low cytotoxicity, making it a valuable candidate for further investigation [84]. The development of selective LMP2 inhibitors presents greater challenges due to the high structural similarity among trypsin-like active sites [79]. Despite this, LU-002i was engineered to enhance selectivity, achieving a 45-fold preference for LMP2 [85]. Interestingly, rapamycin, a macrolide antibiotic, can also selectively inhibit IMPs without affecting the standard proteasome or ubiquitination. Its inhibitory action is partially attributed to suppression of NF-κB (nuclear factor kappa B), pathway activation, and reduced induction of IMPs by inflammatory stimuli [86]. A similar effect has been observed with the natural polyphenol resveratrol [87]. Although new IMP-selective inhibitors continue to be developed, most of these compounds are currently being tested in models of cancer, neurodegenerative, or autoimmune diseases [23,38,73,88,89,90,91].

3. Emerging Insights into the Role of Immunoproteasomes in Chronic Inflammation

IMPs were initially characterized by their enhanced ability to generate antigenic peptides for MHC-I presentation [92]. However, accumulating evidence indicates that their roles extend beyond adaptive immunity, revealing a central function in modulating innate immune responses, regulating cytokine production, and shaping immune cell phenotypes in the setting of chronic inflammation. In models of autoimmune and inflammatory diseases, such as rheumatoid arthritis (RA), systemic lupus erythematosus, inflammatory bowel disease (IBD), and multiple sclerosis, IMPs are markedly upregulated and have been shown to promote inflammation through activation of NLRP3 inflammasomes [93,94], amplifying the production of pro-inflammatory cytokines (e.g., IL-6, IL-17, TNFα), supporting the differentiation of pathogenic Th1 and Th17 cells, and sustaining pro-inflammatory M1 macrophage polarization [35,61,78]. Together, these findings highlight the pro-inflammatory functions of IMPs in chronic inflammation-mediated disorders.
Mechanistically, IMPs regulate the NF-κB signaling pathway, a central regulator of inflammatory responses, immune activation, and cellular survival [95], by promoting the degradation of the IκBα inhibitor, thereby enabling nuclear translocation of NF-κB and the transcription of inflammatory genes [55]. Mice deficient in IMPs display attenuated NF-κB signaling and reduced cytokine production and are protected against autoimmune disease in multiple preclinical models [96]. However, other lines of evidence challenge the view that IMPs are essential for NF-κB activation. For instance, in many cell types, standard proteasomes are sufficient to mediate IκBα degradation and activate NF-κB, even under inflammatory conditions [97]. Moreover, mice deficient in LMP7 or LMP2 retain residual proteasomal activity and often exhibit near-normal NF-κB signaling, suggesting that the constitutive proteasome can functionally compensate for the absence of IMP catalytic subunits [21]. Therefore, while the standard proteasome is clearly indispensable for NF-κB activation, the role of IMPs seems to be more nuanced and context- and cell-type-specific, which is particularly relevant under conditions of chronic inflammation. Rather than acting as primary drivers of NF-κB signaling, IMPs likely function as modulators that influence the magnitude, persistence, and immune consequences of the response.
IMPs are also implicated in the failure to resolve the age-associated, low-grade chronic inflammation that underlies multiple age-related diseases [22,98,99]. Persistent IMP expression in aging tissues may contribute to sustained immune activation and impaired tissue regeneration [100]. In this context, IMPs serve not only as biomarkers of chronic immune activation but also as potential therapeutic targets. As noted above, selective IMP inhibitors, such as ONX 0914, KZR-616, and others, have demonstrated potent anti-inflammatory and immunomodulatory effects in various inflammation models. These findings are summarized in Table 1.
Taken together, these data support a non-redundant, regulatory role for IMPs in the maintenance and amplification of chronic inflammation. Importantly, the development of selective IMP inhibitors has opened new therapeutic avenues. These compounds have shown promising results in preclinical models of chronic inflammatory disorders by dampening pathogenic immune responses while preserving immune surveillance.

4. Immunoproteasomes and Skeletal Muscle Pathologies: A Connection Gaining Recognition

Sarcopenia is an age-associated loss of skeletal muscle mass, strength, and function accompanied by severe adverse health outcomes, such as falls and fractures, functional decline, high health costs, and mortality [2,3]. Despite a considerable body of research, the etiology of sarcopenia remains unclear, and its pathogenesis is still poorly understood. Nevertheless, several major biological processes have been proposed to explain its pathogenesis. These include imbalances in muscle protein turnover [12], neuromuscular junction (NMJ) impairment [112], disturbed mitochondria biogenesis and function [113], insulin resistance [114], fat deposition in muscle [5,115], disturbed myokine production [116], impaired autophagy [117], enhanced muscle cell senescence [118], and non-resolved systemic, chronic, low-grade inflammation [6,119]. Notably, this inflammation has been proposed to link the pathogenesis of sarcopenia with obesity [5,120], type 2 diabetes mellitus [121], and metabolic-dysfunction-associated fatty liver disease (MAFLD) [122], thereby exacerbating sarcopenia manifestations. Altogether, these pathological events result in abnormal muscle fiber reorganization, myofibril degeneration, and myocyte death, as detailed in a series of reviews (e.g., [6,123,124,125,126,127]).
Although extensive research has been undertaken, no pharmacological therapy has yet proven effective for sarcopenia [128,129,130]. At present, the most beneficial strategies for mitigating its progression remain structured exercise programs, particularly resistance training, combined with targeted dietary interventions and supplementation [131]. In elderly individuals, who are often sedentary due to frailty or disability, nutritional management is considered the primary therapeutic option [132]. Among dietary factors, high-quality proteins and the amino acid leucine have shown the greatest promise in supporting skeletal muscle health. Importantly, aging is characterized by reduced muscle sensitivity to the anabolic effects of dietary intake, leading to impaired stimulation of muscle protein synthesis compared with younger adults [133,134,135].
Despite extensive studies in which the role of the UPS in protein degradation in sarcopenia has been repeatedly studied, contradictory findings have emerged, demonstrating, for example, both elevated and unchanged levels of atrogin-1 and MuRF1, two main E3 ubiquitin ligases that play a crucial role in muscle protein breakdown and are key players in muscle atrophy [136,137,138]. Nevertheless, accumulating evidence suggests direct and indirect roles of IMPs in the development of sarcopenia, in close association with chronic inflammation parameters. First, it has been demonstrated that skeletal muscle expresses IMPs; muscle biopsies from older adults revealed an upregulation of IMP subunits, particularly LMP7, which correlates with elevated IL-6 and TNFα expression [24]. Missense mutations in the IMP subunit LMP7 have been reported as the main pathogenic factors in Nakajo–Nishimura syndrome, a rare, inherited autoinflammatory disorder characterized by skeletal muscle atrophy [139,140]. Importantly, this syndrome is also associated with lymphocytic infiltration in the muscles and myositis, thus linking it to inflammation [130]. In a murine model of muscular dystrophy (BlAJ mice) caused by mutations in the dysferlin gene, which leads to muscle weakness and wasting and thus resembles sarcopenia, application of ONX 0914 ameliorated muscle pathology. This improvement was associated with anti-inflammatory M2 macrophage polarization, reduced muscle inflammation, and macrophage-mediated vessel stability, resulting in improved muscle performance in BlAJ mice [27]. In a murine denervation model of sarcopenia, increased LMP7 and MECL-1 expression in skeletal muscle has been found [141], which is suggested not to be directly responsible for muscle wasting but rather to trigger signaling events that ultimately enhance the proteolytic pathways of the cell. In LMP7/MECL-1 double KO (L7M1) mice, no major biochemical, histological, or functional differences were observed in skeletal muscle compared to wild-type controls [142]. However, following exercise, WT muscles upregulated LMP7, while L7M1 muscles did not, suggesting that IMPs may contribute to maintaining skeletal muscle homeostasis.
Recent data suggest a link between skeletal muscle senescence-associated deterioration, chronic inflammation, and IMPs. In mdx mice, a mouse model of human Duchenne muscular dystrophy (DMD), the accumulation of muscle-infiltrating macrophages expressing the senescence-associated secretory phenotype (SASP) release pattern, as well as senescence-related markers (such as p16 and β-gal), has been observed in association with the exhaustion of the skeletal muscle stem cell (satellite cell) pool in dystrophic muscle [143]. Potential involvement of IMPs in muscle cell senescence has been proposed based on studies of 9-month-old mdx mice, in which ONX 0914 administration resulted in an amelioration of the pathological features of muscle atrophy progression. This was associated with a reduced number of macrophages and effector memory T cells in both muscle and the spleen while increasing the number of Tregs and attenuating oxidative stress through improved mitochondrial efficiency [144,145]. Moreover, ONX 0914 mitigates oxidative stress by enhancing mitochondrial efficiency, which in turn leads to a significant reduction in fibrosis, restoration of muscle mechanical function, and improvement in muscle force [26]. Furthermore, these data suggest that the IMP-selective inhibitor ONX 0914 improves the already established and advanced pathological phenotype of aged dystrophic muscle by modulating multiple sarcopenia-associated pathways.
Lipotoxicity, induced by the accumulation of toxic lipid intermediates, such as diacylglycerols, ceramides, and free fatty acids, in skeletal muscle [146,147], is one of the key pathogenic factors in sarcopenic obesity, an age-associated syndrome in which the development of sarcopenia is exacerbated by coexisting obesity [5,148]. Ectopic fatty acids trigger oxidative stress, impair mitochondrial function, enhance insulin resistance [149], and induce production by skeletal muscle myokines, such as myostatin, CCL2, TNFα, IL-1, and IL-6 [150], thus promoting and supporting chronic inflammation [5]. Regarding IMPs, in a study of mice fed a high-fat diet (HFD) and subjected to acute eccentric exercise–induced muscle damage, an increase in skeletal muscle LMP7 and MECL-1 content was observed, along with reduced oxidative stress [151]. These findings suggest that exercise may protect obese individuals from muscle lipotoxicity through IMP upregulation. In another study of HFD-fed mice, although pro-inflammatory cytokines remained unchanged, oxidative protein damage was elevated in the gastrocnemius and tibialis anterior muscles. This intramuscular protein damage coincided with reduced IMP and total proteasome activity, as well as reductions in relative muscle mass, suggesting that proteasome dysregulation may be a critical link between obesity-related oxidative stress and muscle pathology [152]. Notably, LMP7-deficient HFD mice were resistant to obesity and displayed improved glucose tolerance and insulin sensitivity, accompanied by reduced inflammatory responses, such as macrophage infiltration and chemokine expression [153]. Furthermore, in skeletal muscle tissue from marmosets, inhibition of mTOR was associated with increased expression of PSMB5, a core subunit of the 20S proteasome, but not PSMB8, along with elevated expression of mitochondria-targeted protein chaperones [154]. Together, these findings suggest that alterations in IMP activity intersect with oxidative stress, inflammatory responses, and mitochondrial adaptations, thereby providing a mechanistic link between sarcopenia and IMP function.
In addition to sarcopenia, idiopathic inflammatory myopathies (IIMs), a group of complex disorders, are also characterized by significant loss of muscle mass and strength, accompanied by immune cell infiltration into muscle tissue and upregulation of MHC class I expression on myofibers [28]. For example, increased mRNA and protein expression of IMP subunits (PSMB8, PSMB9, and PSMB10) has been detected in muscle biopsies and peripheral blood cells of patients with polymyositis, dermatomyositis, and overlap-syndromes with myositis compared to those with non-inflammatory myopathies and healthy donors [29]. This upregulation correlated with elevated expression of T-cell-specific transcripts in active IIM muscles and was accompanied by increased expression of dendritic cell and monocyte marker genes, suggesting the therapeutic potential of IMP inhibitors for IIMs. Indeed, in a mouse model of polymyositis, administration of ONX 0914 or KZR-616 prevented loss of grip strength and reduced leukocyte infiltration into muscle tissue. Moreover, LMP7-deficient mice were resistant to the induction of polymyositis [30].
In a study of inflamed skeletal muscle tissues from patients with sporadic inclusion body myositis, immune-mediated necrotizing myopathies, and dermatomyositis, two IMP subunits, LMP2 and LMP7, were found to co-localize with MHC-class-I-expressing myofibers [155]. In addition, both myofibers and muscle-infiltrating cells, including CD8+ T cells and CD68+ macrophages, expressed LMP2 and LMP7. Selective inhibition or depletion of LMP7 enhanced TNFα- or IFNγ-mediated expression of cytokines/chemokines (myokines) in myoblasts. Furthermore, specific inhibition of LMP2 or LMP7 reduced IFNγ-induced MHC class I surface expression in myoblasts.
Elevated LMP7 expression was also detected in the muscles of patients with dermatomyositis [156]. In that study, LMP7 overexpression in human skeletal muscle myoblasts (HSMMs) significantly upregulated the muscle atrophy marker MuRF1, type I IFN-related proteins (MxA and IFNβ), and NF-κB pathway-related proteins (pIκBα, pIRF3, and pNF-κBp65). Moreover, LMP7 overexpression markedly reduced HSMM viability, which was partially restored through treatment with the LMP7 inhibitor PR957. In patients with myositis associated with anti-Ku autoantibodies, a rare inflammatory myopathy characterized by prominent upregulation of autophagy, myofiber necrosis, MHC class I and II positivity, and variable endomysial inflammation, conspicuous sarcoplasmic protein aggregates were found to be LMP7-positive [157]. In a murine model of autophagy deficiency, inhibition of eukaryotic translation initiation factor 4E (EIF4E) improved 20S proteasome activity in skeletal muscle, an effect attributed to increased expression of IMP subunits LMP2 and MECL [158]. These findings suggest a regulatory role for IMPs in autophagy within skeletal muscle.

5. Inflammasome Inhibitors in Skeletal Muscle Pathologies: A Double-Edged Sword

Findings discussed in the section above suggest that while the standard proteasome is responsible for degrading myofibrillar proteins during muscle atrophy [137], IMPs may play a role in regulating the inflammatory crosstalk between muscle cells and infiltrating immune cells. Maladaptive immune activation in skeletal muscle pathologies may therefore be exacerbated by IMP-mediated cytokine production and antigen presentation, establishing a vicious cycle of inflammation and muscle wasting [5]. However, IMPs may also protect aged muscle by maintaining proteostasis and supporting myogenesis, particularly during regeneration [25]. Thus, the impact of IMPs in skeletal muscle pathologies appears to be context dependent; they may be protective when facilitating muscle differentiation and protein quality control but detrimental when driving chronic inflammation. This dual role makes them a promising, yet potentially risky, therapeutic target in skeletal muscle pathologies, depending on the timing, disease stage, and balance between proteostatic maintenance and immune activation. This duality raises an important, yet unresolved, question about the rationale for using IMP inhibitors in skeletal muscle pathologies. In other words, “To use or not to use IMP inhibitors in skeletal muscle pathologies—that is the question”.
Why might IMP inhibitors be beneficial? (i) Selective IMP inhibitors (e.g., targeting LMP7 or LMP2) do not shut down all proteasome activity. The constitutive proteasome remains functional, allowing for baseline protein degradation [90,159]. (ii) IMP inhibitors suppress overactive immune signaling without fully blocking proteostasis—reducing cytokine production, antigen presentation, and immune cell infiltration [23]. (iii) IMP inhibition decreases Th1 and Th17 cell differentiation—key drivers of chronic inflammation [62]), limits activation of autoreactive CD8+ T cells [22], and dampens excessive macrophage and dendritic cell cytokine production [160]. (iv) This selective immune modulation can reduce muscle inflammation, as demonstrated in models of IIMs [30], DMD [134], and even systemic inflammation [75].
What might be the detrimental consequences of the application of IMP inhibitors in skeletal muscle pathologies? (i) Inhibiting IMP activity can impair the clearance of damaged proteins, increase proteotoxic stress, and disrupt mitochondrial function, especially in aged or metabolically compromised muscle [72,73,161,162]. (ii) Inhibiting IMP activity can impair the regulation of muscle–immune cell crosstalk, helping to balance pro- and anti-inflammatory signals, leading to suppression of essential immune surveillance, and reduce satellite cell activation, ultimately hindering muscle repair [26,145,163].
Thus, while selective IMP inhibitors may offer benefits by dampening chronic inflammation, their impact on muscle regeneration and tissue homeostasis is context-dependent. In skeletal muscle pathologies, where muscle plasticity is already compromised, untargeted or poorly timed IMP inhibition may worsen outcomes. This underscores the importance of careful therapeutic timing, individualized patient stratification, and a clear understanding of disease stage when considering IMP-based interventions.
To overcome the potential negative effects of IMP inhibitors, combination therapy may offer a promising solution. This approach has been validated in several preclinical models of autoimmune disease [164]. Specifically, the combined inhibition of LMP7 and LMP2, using the selective LMP7 inhibitor PRN1126 alongside LMP2 inhibitors, such as LU-001i or ML604440, was shown to reduce MHC-I surface expression on splenocytes, suppress IL-6 secretion, impair Th17 differentiation, and alleviate symptoms in models of experimental colitis and experimental autoimmune encephalomyelitis (EAE) [163]. Moreover, in a mouse model of anti-collagen antibody-induced arthritis, monotherapy with selective single-site inhibitors had limited efficacy, while the combination of LMP7 with MECL-1 or LMP2 inhibitors produced a clear therapeutic benefit. Notably, the combination of LMP2/MECL-1 with LMP7 inhibition is also effective in constitutive proteasome-resistance cells [165]. Collectively, these findings suggest that effective modulation of IMP activity in inflammatory conditions may require the simultaneous targeting of multiple catalytic subunits. Although data on IMP inhibitor combinations in skeletal muscle-associated disorders are currently lacking, the central role of chronic inflammation in the pathogenesis of skeletal muscle pathologies supports the rationale for exploring such combinatorial strategies in skeletal muscle pathologies.

6. Concluding Remarks

Skeletal muscle pathologies are characterized by progressive muscle wasting that is strongly influenced by age-related chronic inflammation. Within this context, IMPs have emerged as intriguing but paradoxical regulators of muscle homeostasis. Mounting evidence suggests that IMPs may exert both protective and detrimental effects depending on the biological context, disease stage, and cellular environment. On the one hand, IMPs support protein homeostasis and contribute to myogenic differentiation, especially under conditions of cellular stress or regeneration. Their role in maintaining antigen processing and modulating cytokine production may also serve beneficial immune surveillance functions in aging tissue. On the other hand, persistent IMP activation, common in chronic inflammatory states, can exacerbate muscle degradation by amplifying immune responses, sustaining cytokine production, and facilitating maladaptive interactions between muscle cells and infiltrating immune populations. This duality underpins the current uncertainty surrounding the therapeutic use of IMP inhibitors in skeletal muscle pathologies.
Preclinical studies using selective IMP inhibitors (e.g., ONX 0914, KZR-616) in models of autoimmune myositis or neurogenic atrophy have shown promising anti-inflammatory effects and muscle-sparing properties. However, these findings are not easily translatable to skeletal muscle pathologies, where muscle plasticity is already diminished and regenerative capacity is impaired. Inhibiting IMPs in this setting might further compromise proteostasis, impair satellite cell activation, and disrupt immune–satellite cell crosstalk essential for muscle repair. Moreover, unresolved questions remain about the timing, specificity, and combinatorial targeting of IMP subunits. Some studies suggest that selective dual inhibition (e.g., LMP7 and LMP2) yields superior therapeutic efficacy in inflammatory models compared to single-subunit inhibition. Whether such approaches can be safely and effectively applied to skeletal muscle pathologies is unknown and warrants focused investigation.
In summary, the role of IMPs in skeletal muscle pathologies is complex and context-dependent. Their inhibition holds therapeutic promise but may also pose risks to muscle integrity and regeneration. A deeper understanding of IMP function across different stages of muscle aging and inflammation is urgently needed to develop rational, targeted interventions. Future studies should prioritize patient stratification, biomarker development, and integrative approaches that balance immune modulation with preservation of muscle health.
In summary, the role of IMPs in skeletal muscle pathologies is complex and context-dependent. Their inhibition holds therapeutic promise but may also pose risks to muscle integrity and regeneration. As illustrated in Figure 2, a deeper understanding of IMP function across different stages of muscle aging and inflammation will be essential to develop rational, targeted interventions. Future studies should prioritize patient stratification, biomarker development, and integrative approaches that balance immune modulation with the preservation of muscle health.

Author Contributions

A.K. and G.L. contributed to the study’s conception and design. A.K. and G.L. wrote the first draft of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

The study was supported by the Ariel University Research and Development Department (Grant Number: RA2000000457), which supported G.L.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No data were used for the research described in the article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

CCL2, CC motif chemokine ligand 2; CVD, cardiovascular disease; DMD, Duchenne muscular dystrophy; EAE, experimental autoimmune encephalomyelitis; IBD, inflammatory bowel disease; IFN, interferon; INF-κB, nuclear factor kappa B; IIMs, idiopathic inflammatory myopathies; IL, interleukin; IMP, immunoproteasome; HFD, high-fat diet; LMP2, low molecular mass polypeptide 2; MECL1, multicatalytic endo-peptidase complex–like; MHC-I, major histocompatibility complex class; MuRF1, muscle RING-finger protein 1; MxA, myxovirus resistance protein A; NMJ, neuromuscular junction; PBMC, peripheral blood mononuclear cells; pIκBα, phosphorylated inhibitor of kappa light polypeptide gene enhancer in B-cells, alpha pIRF3, phosphorylated interferon regulatory factor 3; POMP, proteasome maturation protein; PSMB9, proteasome subunit beta type-9; RA, rheumatoid arthritis; SASP, senescence-associated secretory phenotype; TLR, toll-like receptor; Th, T helper cell; Treg, regulatory T cell; UPS, ubiquitin–proteasome system.

References

  1. Shafiee, G.; Keshtkar, A.; Soltani, A.; Ahadi, Z.; Larijani, B.; Heshmat, R. Prevalence of sarcopenia in the world: A systematic review and meta- analysis of general population studies. J. Diabetes Metab. Disord. 2017, 16, 21. [Google Scholar] [CrossRef]
  2. Sayer, A.A.; Cruz-Jentoft, A. Sarcopenia definition, diagnosis and treatment: Consensus is growing. Age Ageing 2022, 51, afac220. [Google Scholar] [CrossRef]
  3. Coletta, G.; Phillips, S.M. An elusive consensus definition of sarcopenia impedes research and clinical treatment: A narrative review. Ageing Res. Rev. 2023, 86, 101883. [Google Scholar] [CrossRef]
  4. Bilski, J.; Pierzchalski, P.; Szczepanik, M.; Bonior, J.; Zoladz, J.A. Multifactorial Mechanism of Sarcopenia and Sarcopenic Obesity. Role of Physical Exercise, Microbiota and Myokines. Cells 2022, 11, 160. [Google Scholar] [CrossRef] [PubMed]
  5. Kalinkovich, A.; Livshits, G. Sarcopenic obesity or obese sarcopenia: A cross talk between age-associated adipose tissue and skeletal muscle inflammation as a main mechanism of the pathogenesis. Ageing Res. Rev. 2017, 35, 200–221. [Google Scholar] [CrossRef] [PubMed]
  6. Livshits, G.; Kalinkovich, A. Inflammaging as a common ground for the development and maintenance of sarcopenia, obesity, cardiomyopathy and dysbiosis. Ageing Res. Rev. 2019, 56, 100980. [Google Scholar] [CrossRef] [PubMed]
  7. Liang, Z.; Zhang, T.; Liu, H.; Li, Z.; Peng, L.; Wang, C.; Wang, T. Inflammaging: The ground for sarcopenia? Exp. Gerontol. 2022, 168, 111931. [Google Scholar] [CrossRef]
  8. Antuña, E.; Cachán-Vega, C.; Bermejo-Millo, J.C.; Potes, Y.; Caballero, B.; Vega-Naredo, I.; Coto-Montes, A.; Garcia-Gonzalez, C. Inflammaging: Implications in Sarcopenia. Int. J. Mol. Sci. 2022, 23, 15039. [Google Scholar] [CrossRef]
  9. Jimenez-Gutierrez, G.E.; Martínez-Gómez, L.E.; Martínez-Armenta, C.; Pineda, C.; Martínez-Nava, G.A.; Lopez-Reyes, A. Molecular Mechanisms of Inflammation in Sarcopenia: Diagnosis and Therapeutic Update. Cells 2022, 11, 2359. [Google Scholar] [CrossRef]
  10. Sun, C.; Shen, L.; Zhang, Z.; Xie, X. Therapeutic Strategies for Duchenne Muscular Dystrophy: An Update. Genes 2020, 11, 837. [Google Scholar] [CrossRef]
  11. Dourado, E.; Bottazzi, F.; Cardelli, C.; Conticini, E.; Schmidt, J.; Cavagna, L.; Barsotti, S. Idiopathic inflammatory myopathies: One year in review 2022. Clin. Exp. Rheumatol. 2023, 41, 199–213. [Google Scholar] [CrossRef]
  12. Kim, J.W.; Kim, R.; Choi, H.; Lee, S.J.; Bae, G.U. Understanding of sarcopenia: From definition to therapeutic strategies. Arch. Pharmacal Res. 2021, 44, 876–889. [Google Scholar] [CrossRef]
  13. Brook, M.S.; Wilkinson, D.J.; Atherton, P.J. An update on nutrient modulation in the management of disease-induced muscle wasting: Evidence from human studies. Curr. Opin. Clin. Nutr. Metab. Care 2020, 23, 174–180. [Google Scholar] [CrossRef]
  14. Bonaldo, P.; Sandri, M. Cellular and molecular mechanisms of muscle atrophy. Dis. Model. Mech. 2013, 6, 25–39. [Google Scholar] [CrossRef] [PubMed]
  15. Bhattacharyya, S.; Yu, H.; Mim, C.; Matouschek, A. Regulated protein turnover: Snapshots of the proteasome in action. Nat. Rev. Mol. Cell Biol. 2014, 15, 122–133. [Google Scholar] [CrossRef] [PubMed]
  16. Greene, E.R.; Dong, K.C.; Martin, A. Understanding the 26S proteasome molecular machine from a structural and conformational dynamics perspective. Curr. Opin. Struct. Biol. 2020, 61, 33–41. [Google Scholar] [CrossRef] [PubMed]
  17. Sahu, I.; Glickman, M.H. Structural Insights into Substrate Recognition and Processing by the 20S Proteasome. Biomolecules 2021, 11, 148. [Google Scholar] [CrossRef]
  18. Kandel, R.; Jung, J.; Neal, S. Proteotoxic stress and the ubiquitin proteasome system. Semin. Cell Dev. Biol. 2024, 156, 107–120. [Google Scholar] [CrossRef]
  19. Aki, M.; Shimbara, N.; Takashina, M.; Akiyama, K.; Kagawa, S.; Tamura, T.; Tanahashi, N.; Yoshimura, T.; Tanaka, K.; Ichihara, A. Interferon-gamma induces different subunit organizations and functional diversity of proteasomes. J. Biochem. 1994, 115, 257–269. [Google Scholar] [CrossRef]
  20. Griffin, T.A.; Nandi, D.; Cruz, M.; Fehling, H.J.; Kaer, L.V.; Monaco, J.J.; Colbert, R.A. Immunoproteasome assembly: Cooperative incorporation of interferon gamma (IFN-gamma)-inducible subunits. J. Exp. Med. 1998, 187, 97–104. [Google Scholar] [CrossRef]
  21. Groettrup, M.; Kirk, C.J.; Basler, M. Proteasomes in immune cells: More than peptide producers? Nat. Rev. Immunol. 2010, 10, 73–78. [Google Scholar] [CrossRef]
  22. van den Eshof, B.L.; Medfai, L.; Nolfi, E.; Wawrzyniuk, M.; Sijts, A.J.A.M. The Function of Immunoproteasomes-An Immunologists’ Perspective. Cells 2021, 10, 3360. [Google Scholar] [CrossRef]
  23. Basler, M.; Groettrup, M. On the Role of the Immunoproteasome in Protein Homeostasis. Cells 2021, 10, 3216. [Google Scholar] [CrossRef]
  24. Husom, A.D.; Peters, E.A.; Kolling, E.A.; Fugere, N.A.; Thompson, L.V.; Ferrington, D.A. Altered proteasome function and subunit composition in aged muscle. Arch. Biochem. Biophys. 2004, 421, 67–76. [Google Scholar] [CrossRef]
  25. Cui, Z.; Hwang, S.M.; Gomes, A.V. Identification of the immunoproteasome as a novel regulator of skeletal muscle differentiation. Mol. Cell. Biol. 2014, 34, 96–109. [Google Scholar] [CrossRef]
  26. Zhang, Y.; Hong, S.; Zhang, F.; Yao, K.; Jin, S.; Gao, S.; Liu, Y.; Li, Y.; Zhang, C. Immunoproteasome subunit PSMB8 promotes skeletal muscle regeneration by regulating macrophage phenotyping switch in mice. Am. J. Physiol. Physiol. 2025, 328, C1716–C1729. [Google Scholar] [CrossRef]
  27. Farini, A.; Tripodi, L.; Villa, C.; Napolitano, F.; Strati, F.; Molinaro, D.; Facciotti, F.; Cassani, B.; Torrente, Y. Inhibition of the immunoproteasome modulates innate immunity to ameliorate muscle pathology of dysferlin-deficient BlAJ mice. Cell Death Dis. 2022, 13, 975. [Google Scholar] [CrossRef] [PubMed]
  28. Connolly, C.M.; Gupta, L.; Fujimoto, M.; Machado, P.M.; Paik, J.J. Idiopathic inflammatory myopathies: Current insights and future frontiers. Lancet Rheumatol. 2024, 6, e115–e127. [Google Scholar] [CrossRef] [PubMed]
  29. Ghannam, K.; Martinez-Gamboa, L.; Spengler, L.; Krause, S.; Smiljanovic, B.; Bonin, M.; Bhattarai, S.; Grützkau, A.; Burmester, G.-R.; Häupl, T.; et al. Upregulation of immunoproteasome subunits in myositis indicates active inflammation with involvement of antigen presenting cells, CD8 T-cells and IFNΓ. PLoS ONE 2014, 9, e104048. [Google Scholar] [CrossRef] [PubMed]
  30. Del Rio Oliva, M.; Kirk, C.J.; Groettrup, M.; Basler, M. Effective therapy of polymyositis in mice via selective inhibition of the immunoproteasome. Eur. J. Immunol. 2022, 52, 1510–1522. [Google Scholar] [CrossRef]
  31. Dahlmann, B. Role of proteasomes in disease. BMC Biochem. 2007, 8 (Suppl. 1), S3. [Google Scholar] [CrossRef]
  32. Rousseau, A.; Bertolotti, A. Regulation of proteasome assembly and activity in health and disease. Nat. Rev. Mol. Cell Biol. 2018, 19, 697–712. [Google Scholar] [CrossRef] [PubMed]
  33. Thibaudeau, T.A.; Smith, D.M. A Practical Review of Proteasome Pharmacology. Pharmacol. Rev. 2019, 71, 170–197. [Google Scholar] [CrossRef]
  34. McCarthy, M.K.; Weinberg, J.B. The immunoproteasome and viral infection: A complex regulator of inflammation. Front. Microbiol. 2015, 6, 21. [Google Scholar] [CrossRef] [PubMed]
  35. Basler, M.; Mundt, S.; Bitzer, A.; Schmidt, C.; Groettrup, M. The immunoproteasome: A novel drug target for autoimmune diseases. Clin. Exp. Rheumatol. 2015, 33 (Suppl. 92), S74–S79. [Google Scholar]
  36. Arimochi, H.; Sasaki, Y.; Kitamura, A.; Yasutomo, K. Dysfunctional immunoproteasomes in autoinflammatory diseases. Inflamm. Regen. 2016, 36, 13. [Google Scholar] [CrossRef] [PubMed]
  37. Ettari, R.; Previti, S.; Bitto, A.; Grasso, S.; Zappalà, M. Immunoproteasome-Selective Inhibitors: A Promising Strategy to Treat Hematologic Malignancies, Autoimmune and Inflammatory Diseases. Curr. Med. Chem. 2016, 23, 1217–1238. [Google Scholar] [CrossRef]
  38. Malek, N.; Gladysz, R.; Stelmach, N.; Drag, M. Targeting Microglial Immunoproteasome: A Novel Approach in Neuroinflammatory-Related Disorders. ACS Chem. Neurosci. 2024, 15, 2532–2544. [Google Scholar] [CrossRef]
  39. Coux, O.; Zieba, B.A.; Meiners, S. The Proteasome System in Health and Disease. Adv. Exp. Med. Biol. 2020, 1233, 55–100. [Google Scholar]
  40. Collins, G.A.; Goldberg, A.L. The Logic of the 26S Proteasome. Cell 2017, 169, 792–806. [Google Scholar] [CrossRef]
  41. Bard, J.A.M.; Goodall, E.A.; Greene, E.R.; Jonsson, E.; Dong, K.C.; Martin, A. Structure and Function of the 26S Proteasome. Annu Rev Biochem. 2018, 87, 697–724. [Google Scholar] [CrossRef]
  42. Macagno, A.; Gilliet, M.; Sallusto, F.; Lanzavecchia, A.; Nestle, F.O.; Groettrup, M. Dendritic cells up-regulate immunoproteasomes and the proteasome regulator PA28 during maturation. Eur. J. Immunol. 1999, 29, 4037–4042. [Google Scholar] [CrossRef]
  43. Shin, E.-C.; Seifert, U.; Kato, T.; Rice, C.M.; Feinstone, S.M.; Kloetzel, P.M.; Rehermann, B. Virus-induced type I IFN stimulates generation of immunoproteasomes at the site of infection. J. Clin. Investig. 2006, 116, 3006–3014. [Google Scholar] [CrossRef]
  44. Reis, J.; Guan, X.Q.; Kisselev, A.F.; Papasian, C.J.; Qureshi, A.A.; Morrison, D.C.; Van Way, C.W., 3rd; Vogel, S.N.; Qureshi, N. LPS-induced formation of immunoproteasomes: TNF-α and nitric oxide production are regulated by altered composition of proteasome-active sites. Cell Biochem Biophys. 2011, 60, 77–88. [Google Scholar] [CrossRef] [PubMed]
  45. Pickering, A.M.; Davies, K.J. Degradation of damaged proteins: The main function of the 20S proteasome. Prog. Mol. Biol. Transl. Sci. 2012, 109, 227–248. [Google Scholar]
  46. Fehling, H.J.; Swat, W.; Laplace, C.; Kühn, R.; Rajewsky, K.; Müller, U.; von Boehmer, H. MHC class I expression in mice lacking the proteasome subunit LMP-7. Science 1994, 265, 1234–1237. [Google Scholar] [CrossRef]
  47. Basler, M.; Moebius, J.; Elenich, L.; Groettrup, M.; Monaco, J.J. An altered T cell repertoire in MECL-1-deficient mice. J. Immunol. 2006, 176, 6665–6672. [Google Scholar] [CrossRef]
  48. Van Kaer, L.; Ashton-Rickardt, P.G.; Eichelberger, M.; Gaczynska, M.; Nagashima, K.; Rock, K.L.; Goldberg, A.L.; Doherty, P.C.; Tonegawa, S. Altered peptidase and viral-specific T cell response in LMP2 mutant mice. Immunity 1994, 1, 533–541. [Google Scholar] [CrossRef] [PubMed]
  49. Krüger, E.; Kloetzel, P.M. Immunoproteasomes at the interface of innate and adaptive immune responses: Two faces of one enzyme. Curr. Opin. Immunol. 2012, 24, 77–83. [Google Scholar] [CrossRef] [PubMed]
  50. Heink, S.; Ludwig, D.; Kloetzel, P.M.; Krüger, E. IFN-gamma-induced immune adaptation of the proteasome system is an accelerated and transient response. Proc. Natl. Acad. Sci. USA 2005, 102, 9241–9246. [Google Scholar] [CrossRef]
  51. Hirano, Y.; Kaneko, T.; Okamoto, K.; Bai, M.; Yashiroda, H.; Furuyama, K.; Kato, K.; Tanaka, K.; Murata, S. Dissecting beta-ring assembly pathway of the mammalian 20S proteasome. EMBO J. 2008, 27, 2204–2213. [Google Scholar] [CrossRef]
  52. De, M.; Jayarapu, K.; Elenich, L.; Monaco, J.J.; Colbert, R.A.; Griffin, T.A. Beta 2 subunit propeptides influence cooperative proteasome assembly. J. Biol. Chem. 2003, 278, 6153–6159. [Google Scholar] [CrossRef]
  53. Kingsbury, D.J.; Griffin, T.A.; Colbert, R.A. Novel propeptide function in 20 S proteasome assembly influences beta subunit composition. J. Biol. Chem. 2000, 275, 24156–24162. [Google Scholar] [CrossRef]
  54. Habib, J.A.; Lesenfants, J.; Vigneron, N.; Van den Eynde, B.J. Functional Differences between Proteasome Subtypes. Cells 2022, 11, 421. [Google Scholar] [CrossRef]
  55. Seifert, U.; Bialy, L.P.; Ebstein, F.; Bech-Otschir, D.; Voigt, A.; Schröter, F.; Prozorovski, T.; Lange, N.; Steffen, J.; Rieger, M.; et al. Immunoproteasomes preserve protein homeostasis upon interferon-induced oxidative stress. Cell 2010, 142, 613–624. [Google Scholar] [CrossRef] [PubMed]
  56. Nathan, J.A.; Spinnenhirn, V.; Schmidtke, G.; Basler, M.; Groettrup, M.; Goldberg, A.L. Immuno- and constitutive proteasomes do not differ in their abilities to degrade ubiquitinated proteins. Cell 2013, 152, 1184–1194. [Google Scholar] [CrossRef] [PubMed]
  57. Inholz, K.; Anderl, J.L.; Klawitter, M.; Goebel, H.; Maurits, E.; Kirk, C.J.; Fan, R.A.; Basler, M. Proteasome composition in immune cells implies special immune-cell-specific immunoproteasome function. Eur. J. Immunol. 2024, 54, e2350613. [Google Scholar] [CrossRef]
  58. Moebius, J.; Broek, M.; Groettrup, M.; Basler, M. Immunoproteasomes are essential for survival and expansion of T cells in virus-infected mice. Eur. J. Immunol. 2010, 40, 3439–3449. [Google Scholar] [CrossRef] [PubMed]
  59. Caudill, C.M.; Jayarapu, K.; Elenich, L.; Monaco, J.J.; Colbert, R.A.; Griffin, T.A. T cells lacking immunoproteasome subunits MECL-1 and LMP7 hyperproliferate in response to polyclonal mitogens. J. Immunol. 2006, 176, 4075–4082. [Google Scholar] [CrossRef]
  60. Kalim, K.W.; Basler, M.; Kirk, C.J.; Groettrup, M. Immunoproteasome subunit LMP7 deficiency and inhibition suppresses Th1 and Th17 but enhances regulatory T cell differentiation. J. Immunol. 2012, 189, 4182–4193. [Google Scholar] [CrossRef]
  61. Vachharajani, N.; Joeris, T.; Luu, M.; Hartmann, S.; Pautz, S.; Jenike, E.; Pantazis, G.; Prinz, I.; Hofer, M.J.; Steinhoff, U.; et al. Prevention of colitis-associated cancer by selective targeting of immunoproteasome subunit LMP7. Oncotarget 2017, 8, 50447–50459. [Google Scholar] [CrossRef]
  62. Oliveri, F.; Mink, D.; Muchamuel, T.; Basler, M. Immunoproteasome Inhibition Impairs Differentiation but Not Survival of T Helper 17 Cells. Cells 2025, 14, 689. [Google Scholar] [CrossRef]
  63. Chen, S.; Kammerl, I.E.; Vosyka, O.; Baumann, T.; Yu, Y.; Wu, Y.; Irmler, M.; Overkleeft, H.S.; Beckers, J.; Eickelberg, O.; et al. Immunoproteasome dysfunction augments alternative polarization of alveolar macrophages. Cell Death Differ. 2016, 23, 1026–1037. [Google Scholar] [CrossRef] [PubMed]
  64. Liao, J.; Xie, Y.; Lin, Q.; Yang, X.; An, X.; Xia, Y.; Du, J.; Wang, F.; Li, H.H. Immunoproteasome subunit β5i regulates diet-induced atherosclerosis through altering MERTK-mediated efferocytosis in Apoe knockout mice. J. Pathol. 2020, 250, 275–287. [Google Scholar] [CrossRef] [PubMed]
  65. Kremer, M.; Henn, A.; Kolb, C.; Basler, M.; Moebius, J.; Guillaume, B.; Leist, M.; Van den Eynde, B.J.; Groettrup, M. Reduced immunoproteasome formation and accumulation of immunoproteasomal precursors in the brains of lymphocytic choriomeningitis virus-infected mice. J. Immunol. 2010, 185, 5549–5560. [Google Scholar] [CrossRef]
  66. Mundt, S.; Engelhardt, B.; Kirk, C.J.; Groettrup, M.; Basler, M. Inhibition and deficiency of the immunoproteasome subunit LMP7 attenuates LCMV-induced meningitis. Eur. J. Immunol. 2016, 46, 104–113. [Google Scholar] [CrossRef] [PubMed]
  67. Basler, M.; Li, J.; Groettrup, M. On the role of the immunoproteasome in transplant rejection. Immunogenetics 2019, 71, 263–271. [Google Scholar] [CrossRef]
  68. Keller, I.E.; Vosyka, O.; Takenaka, S.; Kloß, A.; Dahlmann, B.; Willems, L.I.; Verdoes, M.; Overkleeft, H.S.; Marcos, E.; Adnot, S.; et al. Regulation of immunoproteasome function in the lung. Sci. Rep. 2015, 5, 10230. [Google Scholar] [CrossRef]
  69. Kammerl, I.E.; Dann, A.; Mossina, A.; Brech, D.; Lukas, C.; Vosyka, O.; Nathan, P.; Conlon, T.M.; Wagner, D.E.; Overkleeft, H.S.; et al. Impairment of Immunoproteasome Function by Cigarette Smoke and in Chronic Obstructive Pulmonary Disease. Am. J. Respir. Crit. Care Med. 2016, 193, 1230–1241. [Google Scholar] [CrossRef]
  70. Desterke, C.; Turhan, A.G.; Bennaceur-Griscelli, A.; Griscelli, F. HLA-dependent heterogeneity and macrophage immunoproteasome activation during lung COVID-19 disease. J. Transl. Med. 2021, 19, 290. [Google Scholar] [CrossRef]
  71. Davidson, K.; Pickering, A.M. The proteasome: A key modulator of nervous system function, brain aging, and neurodegenerative disease. Front. Cell Dev. Biol. 2023, 11, 1124907. [Google Scholar] [CrossRef]
  72. Church, T.R.; Margolis, S.S. Mechanisms of ubiquitin-independent proteasomal degradation and their roles in age-related neurodegenerative disease. Front. Cell Dev. Biol. 2025, 12, 1531797. [Google Scholar] [CrossRef]
  73. Nie, Y.; Ma, Z.; Zhang, B.; Sun, M.; Zhang, D.; Li, H.H.; Song, X. The role of the immunoproteasome in cardiovascular disease. Pharmacol. Res. 2024, 204, 107215. [Google Scholar] [CrossRef]
  74. Ott, C. Mapping the interplay of immunoproteasome and autophagy in different heart failure phenotypes. Free. Radic. Biol. Med. 2024, 218, 149–165. [Google Scholar] [CrossRef] [PubMed]
  75. Xi, J.; Zhuang, R.; Kong, L.; He, R.; Zhu, H.; Zhang, J. Immunoproteasome-selective inhibitors: An overview of recent developments as potential drugs for hematologic malignancies and autoimmune diseases. Eur. J. Med. Chem. 2019, 182, 111646. [Google Scholar] [CrossRef] [PubMed]
  76. Abramson, H.N. Immunotherapy of Multiple Myeloma: Current Status as Prologue to the Future. Int. J. Mol. Sci. 2023, 24, 15674. [Google Scholar] [CrossRef]
  77. Arastu-Kapur, S.; Anderl, J.L.; Kraus, M.; Parlati, F.; Shenk, K.D.; Lee, S.J.; Muchamuel, T.; Bennett, M.K.; Driessen, C.; Ball, A.J.; et al. Nonproteasomal targets of the proteasome inhibitors bortezomib and carfilzomib: A link to clinical adverse events. Clin. Cancer Res. 2011, 17, 2734–2743. [Google Scholar] [CrossRef]
  78. Muchamuel, T.; Basler, M.; Aujay, M.A.; Suzuki, E.; Kalim, K.W.; Lauer, C.; Sylvain, C.; Ring, E.R.; Shields, J.; Jiang, J.; et al. A selective inhibitor of the immunoproteasome subunit LMP7 blocks cytokine production and attenuates progression of experimental arthritis. Nat. Med. 2009, 15, 781–787. [Google Scholar] [CrossRef]
  79. Huber, E.M.; Basler, M.; Schwab, R.; Heinemeyer, W.; Kirk, C.J.; Groettrup, M.; Groll, M. Immuno- and constitutive proteasome crystal structures reveal differences in substrate and inhibitor specificity. Cell 2012, 148, 727–738. [Google Scholar] [CrossRef]
  80. Kisselev, A.F. Site-Specific Proteasome Inhibitors. Biomolecules 2021, 12, 54. [Google Scholar] [CrossRef] [PubMed]
  81. Muchamuel, T.; Fan, R.A.; Anderl, J.L.; Bomba, D.J.; Johnson, H.W.B.; Lowe, E.; Tuch, B.B.; McMinn, D.L.; Millare, B.; Kirk, C.J. Zetomipzomib (KZR-616) attenuates lupus in mice via modulation of innate and adaptive immune responses. Front. Immunol. 2023, 14, 1043680. [Google Scholar] [CrossRef]
  82. Klein, M.; Busch, M.; Friese-Hamim, M.; Crosignani, S.; Fuchss, T.; Musil, D.; Rohdich, F.; Sanderson, M.P.; Seenisamy, J.; Walter-Bausch, G.; et al. Structure-Based Optimization and Discovery of M3258, a Specific Inhibitor of the Immunoproteasome Subunit LMP7 (β5i). J. Med. Chem. 2021, 64, 10230–10245. [Google Scholar] [CrossRef] [PubMed]
  83. Karreci, E.S.; Fan, H.; Uehara, M.; Mihali, A.B.; Singh, P.K.; Kurdi, A.T.; Solhjou, Z.; Riella, L.V.; Ghobrial, I.; Laragione, T.; et al. Brief treatment with a highly selective immunoproteasome inhibitor promotes long-term cardiac allograft acceptance in mice. Proc. Natl. Acad. Sci. USA 2016, 113, E8425–E8432. [Google Scholar] [CrossRef] [PubMed]
  84. Kuhn, D.J.; Hunsucker, S.A.; Chen, Q.; Voorhees, P.M.; Orlowski, M.; Orlowski, R.Z. Targeted inhibition of the immunoproteasome is a potent strategy against models of multiple myeloma that overcomes resistance to conventional drugs and nonspecific proteasome inhibitors. Blood 2009, 113, 4667–4676. [Google Scholar] [CrossRef]
  85. Xin, B.-T.; Huber, E.M.; de Bruin, G.; Heinemeyer, W.; Maurits, E.; Espinal, C.; Du, Y.; Janssens, M.; Weyburne, E.S.; Kisselev, A.F.; et al. Structure-Based Design of Inhibitors Selective for Human Proteasome β2c or β2i Subunits. J. Med. Chem. 2019, 62, 1626–1642. [Google Scholar] [CrossRef]
  86. Zhang, H.M.; Fu, J.; Hamilton, R.; Diaz, V.; Zhang, Y. The mammalian target of rapamycin modulates the immunoproteasome system in the heart. J. Mol. Cell. Cardiol. 2015, 86, 158–167. [Google Scholar] [CrossRef]
  87. Chen, C.; Zou, L.X.; Lin, Q.Y.; Yan, X.; Bi, H.L.; Xie, X.; Wang, S.; Wang, Q.S.; Zhang, Y.L.; Li, H.H. Resveratrol as a new inhibitor of immunoproteasome prevents PTEN degradation and attenuates cardiac hypertrophy after pressure overload. Redox Biol. 2019, 20, 390–401. [Google Scholar] [CrossRef]
  88. Tripathi, S.C.; Vedpathak, D.; Ostrin, E.J. The Functional and Mechanistic Roles of Immunoproteasome Subunits in Cancer. Cells 2021, 10, 3587. [Google Scholar] [CrossRef]
  89. Huber, E.M.; Groll, M. A Nut for Every Bolt: Subunit-Selective Inhibitors of the Immunoproteasome and Their Therapeutic Potential. Cells 2021, 10, 1929. [Google Scholar] [CrossRef]
  90. Mancuso, F.; Di Chio, C.; Di Matteo, F.; Smaldone, G.; Iraci, N.; Giofrè, S.V. Recent Advances in the Development of Immunoproteasome Inhibitors as Anti-Cancer Agents: The Past 5 Years. Molecules 2025, 30, 755. [Google Scholar] [CrossRef] [PubMed]
  91. Simon, P.Y.R.; David, R. Alzheimer’s Disease, β-Amyloid Peptides, and Ubiquitin-Proteasome System: Nutritherapeutic Insights. Neurodegener. Dis. 2025, 25, 76–87. [Google Scholar] [CrossRef] [PubMed]
  92. Kloetzel, P.M.; Ossendorp, F. Proteasome and peptidase function in MHC-class-I-mediated antigen presentation. Curr. Opin. Immunol. 2004, 16, 76–81. [Google Scholar] [CrossRef]
  93. Chen, X.; Wang, Y.; Yao, N.; Lin, Z. Immunoproteasome modulates NLRP3 inflammasome-mediated neuroinflammation under cerebral ischaemia and reperfusion conditions. J. Cell. Mol. Med. 2022, 26, 462–474. [Google Scholar] [CrossRef]
  94. Lee, Y.; Yoon, B.; Son, S.; Cho, E.; Kim, K.B.; Choi, E.Y.; Kim, D.E. Inhibition of Immunoproteasome Attenuates NLRP3 Inflammasome Response by Regulating E3 Ubiquitin Ligase TRIM31. Cells 2024, 13, 675. [Google Scholar] [CrossRef]
  95. Kanarek, N.; Ben-Neriah, Y. Regulation of NF-κB by ubiquitination and degradation of the IκBs. Immunol. Rev. 2012, 246, 77–94. [Google Scholar] [CrossRef]
  96. Bitzer, A.; Basler, M.; Krappmann, D.; Groettrup, M. Immunoproteasome subunit deficiency has no influence on the canonical pathway of NF-κB activation. Mol. Immunol. 2017, 83, 147–153. [Google Scholar] [CrossRef] [PubMed]
  97. Lee, K.H.; Jeong, J.; Yoo, C.G. Long-term incubation with proteasome inhibitors (PIs) induces IκBα degradation via the lysosomal pathway in an IκB kinase (IKK)-dependent and IKK-independent manner. J. Biol. Chem. 2013, 288, 32777–32786. [Google Scholar] [CrossRef] [PubMed]
  98. Franceschi, C.; Santoro, A.; Capri, M. The complex relationship between Immunosenescence and Inflammaging: Special issue on the New Biomedical Perspectives. Semin. Immunopathol. 2020, 42, 517–520. [Google Scholar] [CrossRef]
  99. Nathan, C. Nonresolving inflammation redux. Immunity 2022, 55, 592–605. [Google Scholar] [CrossRef]
  100. Johnston-Carey, H.K.; Pomatto, L.C.D.; Davies, K.J.A. The Immunoproteasome in oxidative stress, aging, and disease. Crit. Rev. Biochem. Mol. Biol. 2015, 51, 268–281. [Google Scholar] [CrossRef]
  101. Ichikawa, H.T.; Conley, T.; Muchamuel, T.; Jiang, J.; Lee, S.; Owen, T.; Barnard, J.; Nevarez, S.; Goldman, B.I.; Kirk, C.J.; et al. Beneficial effect of novel proteasome inhibitors in murine lupus via dual inhibition of type I interferon and autoantibody-secreting cells. Arthritis Rheum. 2012, 64, 493–503. [Google Scholar] [CrossRef] [PubMed]
  102. Oliva, M.d.R.; Mellett, M.; Basler, M. Immunoproteasome inhibition attenuates experimental psoriasis. Front. Immunol. 2022, 13, 1075615. [Google Scholar] [CrossRef]
  103. Schaftenaar, F.H.; van Dam, A.D.; de Bruin, G.; Depuydt, M.A.; de Mol, J.; Amersfoort, J.; Douna, H.; Meijer, M.; Kröner, M.J.; van Santbrink, P.J.; et al. Immunoproteasomal Inhibition With ONX-0914 Attenuates Atherosclerosis and Reduces White Adipose Tissue Mass and Metabolic Syndrome in Mice. Arter. Thromb. Vasc. Biol. 2024, 44, 1346–1364. [Google Scholar] [CrossRef]
  104. Zhang, L.; Xia, Q.; Li, W.; Liu, Q.; Zhang, L.; Tian, X.; Ye, L.; Wang, G.; Peng, Q. Immunoproteasome subunit β5i promotes perifascicular muscle atrophy in dermatomyositis by upregulating RIG-I. RMD Open 2023, 9, e002818. [Google Scholar] [CrossRef]
  105. Du, S.H.; Xiang, Y.J.; Liu, L.; Nie, M.; Hou, Y.; Wang, L.; Li, B.B.; Xu, M.; Teng, Q.L.; Peng, J.; et al. Co-Inhibition of the Immunoproteasome Subunits LMP2 and LMP7 Ameliorates Immune Thrombocytopenia. Front. Immunol. 2021, 11, 603278. [Google Scholar] [CrossRef]
  106. Chang, H.H.; Lin, Y.H.; Chen, T.M.; Tsai, Y.L.; Lai, C.R.; Tsai, W.C.; Cheng, Y.C.; Chen, Y. ONX-0914 Induces Apoptosis and Autophagy with p53 Regulation in Human Glioblastoma Cells. Cancers 2022, 14, 5712. [Google Scholar] [CrossRef]
  107. Li, J.; Liu, N.; Zhou, H.; Xian, P.; Song, Y.; Tang, X.; Li, Y.; Basler, M. Immunoproteasome inhibition prevents progression of castration-resistant prostate cancer. Br. J. Cancer 2023, 128, 1377–1390. [Google Scholar] [CrossRef]
  108. Downey-Kopyscinski, S.; Daily, E.W.; Gautier, M.; Bhatt, A.; Florea, B.I.; Mitsiades, C.S.; Richardson, P.G.; Driessen, C.; Overkleeft, H.S.; Kisselev, A.F. An inhibitor of proteasome β2 sites sensitizes myeloma cells to immunoproteasome inhibitors. Blood Adv. 2018, 2, 2443–2451. [Google Scholar] [CrossRef]
  109. Bhattarai, D.; Lee, M.J.; Baek, A.; Yeo, I.J.; Miller, Z.; Baek, Y.M.; Lee, S.; Kim, D.E.; Hong, J.T.; Kim, K.B. LMP2 Inhibitors as a Potential Treatment for Alzheimer’s Disease. J. Med. Chem. 2020, 63, 3763–3783. [Google Scholar] [CrossRef] [PubMed]
  110. Kioon, M.D.A.; Pierides, M.; Pannellini, T.; Lin, G.; Nathan, C.F.; Barrat, F.J. Noncytotoxic Inhibition of the Immunoproteasome Regulates Human Immune Cells In Vitro and Suppresses Cutaneous Inflammation in the Mouse. J. Immunol. 2021, 206, 1631–1641. [Google Scholar] [CrossRef] [PubMed]
  111. Xie, X.; Lee, J.; Manyam, G.C.; Pearson, T.; Walter-Bausch, G.; Friese-Hamim, M.; Zhao, S.; Jabs, J.; Manginelli, A.A.; Piske, N.; et al. LMP7-Specific Inhibitor M3258 Modulates the Tumor Microenvironment of Triple-Negative Breast Cancer and Inflammatory Breast Cancer. Cancers 2025, 17, 1887. [Google Scholar] [CrossRef]
  112. Moallemian, R.; Rehman, A.U.; Zhao, N.; Wang, H.; Chen, H.; Lin, G.; Ma, X.; Yu, J. Immunoproteasome inhibitor DPLG3 attenuates experimental colitis by restraining NF-κB activation. Biochem. Pharmacol. 2020, 177, 113964. [Google Scholar] [CrossRef] [PubMed]
  113. Moreira-Pais, A.; Ferreira, R.; Oliveira, P.A.; Duarte, J.A. A neuromuscular perspective of sarcopenia pathogenesis: Deciphering the signaling pathways involved. Geroscience 2022, 44, 1199–1213. [Google Scholar] [CrossRef]
  114. Marzetti, E.; Calvani, R.; Coelho-Júnior, H.J.; Landi, F.; Picca, A. Mitochondrial Quantity and Quality in Age-Related Sarcopenia. Int. J. Mol. Sci. 2024, 25, 2052. [Google Scholar] [CrossRef]
  115. Liu, Z.-J.; Zhu, C.-F. Causal relationship between insulin resistance and sarcopenia. Diabetol. Metab. Syndr. 2023, 15, 46. [Google Scholar] [CrossRef]
  116. Wang, L.; Valencak, T.G.; Shan, T. Fat infiltration in skeletal muscle: Influential triggers and regulatory mechanism. iScience 2024, 27, 109221. [Google Scholar] [CrossRef]
  117. Gomarasca, M.; Banfi, G.; Lombardi, G. Myokines: The endocrine coupling of skeletal muscle and bone. Adv. Clin. Chem. 2020, 94, 155–218. [Google Scholar]
  118. Xie, G.; Jin, H.; Mikhail, H.; Pavel, V.; Yang, G.; Ji, B.; Lu, B.; Li, Y. Autophagy in sarcopenia: Possible mechanisms and novel therapies. Biomed. Pharmacother. 2023, 165, 115147. [Google Scholar] [CrossRef] [PubMed]
  119. He, Y.; Xie, W.; Li, H.; Jin, H.; Zhang, Y.; Li, Y. Cellular Senescence in Sarcopenia: Possible Mechanisms and Therapeutic Potential. Front. Cell Dev. Biol. 2022, 9, 793088. [Google Scholar] [CrossRef] [PubMed]
  120. Kalinkovich, A.; Becker, M.; Livshits, G. New Horizons in the Treatment of Age-Associated Obesity, Sarcopenia and Osteoporosis. Drugs Aging 2022, 39, 673–683. [Google Scholar] [CrossRef]
  121. Rohm, T.V.; Meier, D.T.; Olefsky, J.M.; Donath, M.Y. Inflammation in obesity, diabetes, and related disorders. Immunity 2022, 55, 31–55. [Google Scholar] [CrossRef]
  122. Izzo, A.; Massimino, E.; Riccardi, G.; Della Pepa, G. A Narrative Review on Sarcopenia in Type 2 Diabetes Mellitus: Prevalence and Associated Factors. Nutrients 2021, 13, 183. [Google Scholar] [CrossRef]
  123. Losasso, M.R.; Parussolo, M.L.C.; Silva, A.O.; Direito, R.; Quesada, K.; Detregiachi, C.R.P.; Bechara, M.D.; Méndez-Sánchez, N.; Abenavoli, L.; Araújo, A.C.; et al. Unraveling the Metabolic Pathways Between Metabolic-Associated Fatty Liver Disease (MAFLD) and Sarcopenia. Int. J. Mol. Sci. 2025, 26, 4673. [Google Scholar] [CrossRef]
  124. Kalinkovich, A.; Livshits, G. Sarcopenia-The search for emerging biomarkers. Ageing Res. Rev. 2015, 22, 58–71. [Google Scholar] [CrossRef]
  125. Larsson, L.; Degens, H.; Li, M.; Salviati, L.; Lee, Y.I.; Thompson, W.; Kirkland, J.L.; Sandri, M. Sarcopenia: Aging-Related Loss of Muscle Mass and Function. Physiol. Rev. 2019, 99, 427–511. [Google Scholar] [CrossRef]
  126. Nishikawa, H.; Fukunishi, S.; Asai, A.; Yokohama, K.; Nishiguchi, S.; Higuchi, K. Pathophysiology and mechanisms of primary sarcopenia (Review). Int. J. Mol. Med. 2021, 48, 156. [Google Scholar] [CrossRef]
  127. Wu, J.; Ding, P.; Wu, H.; Yang, P.; Guo, H.; Tian, Y.; Meng, L.; Zhao, Q. Sarcopenia: Molecular regulatory network for loss of muscle mass and function. Front. Nutr. 2023, 10, 1037200. [Google Scholar] [CrossRef]
  128. Liu, D.; Wang, S.; Liu, S.; Wang, Q.; Che, X.; Wu, G. Frontiers in sarcopenia: Advancements in diagnostics, molecular mechanisms, and therapeutic strategies. Mol. Asp. Med. 2024, 97, 101270. [Google Scholar] [CrossRef] [PubMed]
  129. Cho, M.R.; Lee, S.; Song, S.K. A Review of Sarcopenia Pathophysiology, Diagnosis, Treatment and Future Direction. J. Korean Med. Sci. 2022, 37, e146. [Google Scholar] [CrossRef]
  130. Rolland, Y.; Dray, C.; Vellas, B.; Barreto, P.D.S. Current and investigational medications for the treatment of sarcopenia. Metabolism 2023, 149, 155597. [Google Scholar] [CrossRef] [PubMed]
  131. Najm, A.; Niculescu, A.-G.; Grumezescu, A.M.; Beuran, M. Emerging Therapeutic Strategies in Sarcopenia: An Updated Review on Pathogenesis and Treatment Advances. Int. J. Mol. Sci. 2024, 25, 4300. [Google Scholar] [CrossRef] [PubMed]
  132. Rondanelli, M.; Nichetti, M.; Peroni, G.; Faliva, M.A.; Naso, M.; Gasparri, C.; Perna, S.; Oberto, L.; Di Paolo, E.; Riva, A.; et al. Where to Find Leucine in Food and How to Feed Elderly With Sarcopenia in Order to Counteract Loss of Muscle Mass: Practical Advice. Front. Nutr. 2021, 7, 622391. [Google Scholar] [CrossRef]
  133. Shen, Y.; Shi, Q.; Nong, K.; Li, S.; Yue, J.; Huang, J.; Dong, B.; Beauchamp, M.; Hao, Q. Exercise for sarcopenia in older people: A systematic review and network meta-analysis. J. Cachex. Sarcopenia Muscle 2023, 14, 1199–1211. [Google Scholar] [CrossRef]
  134. Deutz, N.E.; Bauer, J.M.; Barazzoni, R.; Biolo, G.; Boirie, Y.; Bosy-Westphal, A.; Cederholm, T.; Cruz-Jentoft, A.; Krznariç, Z.; Nair, K.S.; et al. Protein intake and exercise for optimal muscle function with aging: Recommendations from the ESPEN Expert Group. Clin. Nutr. 2014, 33, 929–936. [Google Scholar] [CrossRef]
  135. Devries, M.C.; McGlory, C.; Bolster, D.R.; Kamil, A.; Rahn, M.; Harkness, L.; Baker, S.K.; Phillips, S.M. Protein leucine content is a determinant of shorter- and longer-term muscle protein synthetic responses at rest and following resistance exercise in healthy older women: A randomized, controlled trial. Am. J. Clin. Nutr. 2018, 107, 217–226. [Google Scholar] [CrossRef]
  136. Martínez-Arnau, F.M.; Fonfría-Vivas, R.; Buigues, C.; Castillo, Y.; Molina, P.; Hoogland, A.J.; van Doesburg, F.; Pruimboom, L.; Fernández-Garrido, J.; Cauli, O. Effects of Leucine Administration in Sarcopenia: A Randomized and Placebo-controlled Clinical Trial. Nutrients 2020, 12, 932. [Google Scholar] [CrossRef] [PubMed]
  137. Sakuma, K.; Aoi, W.; Yamaguchi, A. Molecular mechanism of sarcopenia and cachexia: Recent research advances. Pflug. Arch. 2017, 469, 573–591. [Google Scholar] [CrossRef]
  138. Haberecht-Müller, S.; Krüger, E.; Fielitz, J. Out of Control: The Role of the Ubiquitin Proteasome System in Skeletal Muscle during Inflammation. Biomolecules 2021, 11, 1327. [Google Scholar] [CrossRef]
  139. Pang, X.; Zhang, P.; Chen, X.; Liu, W. Ubiquitin-proteasome pathway in skeletal muscle atrophy. Front. Physiol. 2023, 14, 1289537. [Google Scholar] [CrossRef] [PubMed]
  140. Arima, K.; Kinoshita, A.; Mishima, H.; Kanazawa, N.; Kaneko, T.; Mizushima, T.; Ichinose, K.; Nakamura, H.; Tsujino, A.; Kawakami, A.; et al. Proteasome assembly defect due to a proteasome subunit beta type 8 (PSMB8) mutation causes the autoinflammatory disorder, Nakajo-Nishimura syndrome. Proc. Natl. Acad. Sci. USA 2011, 108, 14914–14919. [Google Scholar] [CrossRef]
  141. Ayaki, T.; Murata, K.; Kanazawa, N.; Uruha, A.; Ohmura, K.; Sugie, K.; Kasagi, S.; Li, F.; Mori, M.; Nakajima, R.; et al. Myositis with sarcoplasmic inclusions in Nakajo–Nishimura syndrome: A genetic inflammatory myopathy. Neuropathol. Appl. Neurobiol. 2020, 46, 579–587. [Google Scholar] [CrossRef] [PubMed]
  142. Liu, H.M.; Ferrington, D.A.; Baumann, C.W.; Thompson, L.V. Denervation-Induced Activation of the Standard Proteasome and Immunoproteasome. PLoS ONE 2016, 11, e0166831. [Google Scholar] [CrossRef] [PubMed]
  143. Baumann, C.W.; Kwak, D.; Ferrington, D.A.; Thompson, L.V. Downhill exercise alters immunoproteasome content in mouse skeletal muscle. Cell Stress Chaperon. 2018, 23, 507–517. [Google Scholar] [CrossRef] [PubMed]
  144. Liu, L.; Yue, X.; Sun, Z.; Hambright, W.S.; Feng, Q.; Cui, Y.; Huard, J.; Robbins, P.D.; Wang, Z.; Mu, X. Senolytic elimination of senescent macrophages restores muscle stem cell function in severely dystrophic muscle. Aging 2022, 14, 7650–7661. [Google Scholar] [CrossRef]
  145. Farini, A.; Sitzia, C.; Cassani, B.; Cassinelli, L.; Rigoni, R.; Colleoni, F.; Fusco, N.; Gatti, S.; Bella, P.; Villa, C.; et al. Therapeutic Potential of Immunoproteasome Inhibition in Duchenne Muscular Dystrophy. Mol. Ther. 2016, 24, 1898–1912. [Google Scholar] [CrossRef]
  146. Tripodi, L.; Molinaro, D.; Fortunato, F.; Mella, C.; Cassani, B.; Torrente, Y.; Farini, A. Immunoproteasome Inhibition Ameliorates Aged Dystrophic Mouse Muscle Environment. Int. J. Mol. Sci. 2022, 23, 14657. [Google Scholar] [CrossRef]
  147. Buch, A.; Carmeli, E.; Boker, L.K.; Marcus, Y.; Shefer, G.; Kis, O.; Berner, Y.; Stern, N. Muscle function and fat content in relation to sarcopenia, obesity and frailty of old age—An overview. Exp. Gerontol. 2016, 76, 25–32. [Google Scholar] [CrossRef]
  148. Nishi, H.; Higashihara, T.; Inagi, R. Lipotoxicity in Kidney, Heart, and Skeletal Muscle Dysfunction. Nutrients 2019, 11, 1664. [Google Scholar] [CrossRef]
  149. Prado, C.M.; Batsis, J.A.; Donini, L.M.; Gonzalez, M.C.; Siervo, M. Sarcopenic obesity in older adults: A clinical overview. Nat. Rev. Endocrinol. 2024, 20, 261–277. [Google Scholar] [CrossRef] [PubMed]
  150. Li, C.; Yu, K.; Shyh-Chang, N.; Jiang, Z.; Liu, T.; Ma, S.; Luo, L.; Guang, L.; Liang, K.; Ma, W.; et al. Pathogenesis of sarcopenia and the relationship with fat mass: Descriptive review. J. Cachex. Sarcopenia Muscle 2022, 13, 781–794. [Google Scholar] [CrossRef]
  151. Cleasby, M.E.; Jamieson, P.M.; Atherton, P.J. Insulin resistance and sarcopenia: Mechanistic links between common co-morbidities. J. Endocrinol. 2016, 229, R67–R81. [Google Scholar] [CrossRef]
  152. Fletcher, E.; Gordon, P.M. Obesity-induced alterations to the immunoproteasome: A potential link to intramuscular lipotoxicity. Appl. Physiol. Nutr. Metab. 2021, 46, 485–493. [Google Scholar] [CrossRef] [PubMed]
  153. Fletcher, E.; Wiggs, M.; Greathouse, K.L.; Morgan, G.B.; Gordon, P.M. Impaired proteostasis in obese skeletal muscle relates to altered immunoproteasome activity. Appl. Physiol. Nutr. Metab. 2022, 47, 555–564. [Google Scholar] [CrossRef]
  154. Kimura, H.; Usui, F.; Karasawa, T.; Kawashima, A.; Shirasuna, K.; Inoue, Y.; Komada, T.; Kobayashi, M.; Mizushina, Y.; Kasahara, T.; et al. Immunoproteasome subunit LMP7 Deficiency Improves Obesity and Metabolic Disorders. Sci. Rep. 2015, 5, 15883. [Google Scholar] [CrossRef]
  155. Lelegren, M.; Liu, Y.; Ross, C.; Tardif, S.; Salmon, A.B. Pharmaceutical inhibition of mTOR in the common marmoset: Effect of rapamycin on regulators of proteostasis in a non-human primate. Pathobiol. Aging Age-Relat. Dis. 2016, 6, 31793. [Google Scholar] [CrossRef]
  156. Bhattarai, S.; Ghannam, K.; Krause, S.; Benveniste, O.; Marg, A.; de Bruin, G.; Xin, B.T.; Overkleeft, H.S.; Spuler, S.; Stenzel, W.; et al. The immunoproteasomes are key to regulate myokines and MHC class I expression in idiopathic inflammatory myopathies. J. Autoimmun. 2016, 75, 118–129. [Google Scholar] [CrossRef]
  157. Holzer, M.T.; Uruha, A.; Roos, A.; Hentschel, A.; Schänzer, A.; Weis, J.; Claeys, K.G.; Schoser, B.; Montagnese, F.; Goebel, H.-H.; et al. Anti-Ku + myositis: An acquired inflammatory protein-aggregate myopathy. Acta Neuropathol. 2024, 148, 6. [Google Scholar] [CrossRef]
  158. Dong, H.; Lyu, Y.; Huang, C.Y.; Tsai, S.Y. Limiting cap-dependent translation increases 20S proteasomal degradation and protects the proteomic integrity in autophagy-deficient skeletal muscle. Autophagy 2025, 21, 1212–1227. [Google Scholar] [CrossRef]
  159. Miller, Z.; Ao, L.; Kim, K.B.; Lee, W. Inhibitors of the immunoproteasome: Current status and future directions. Curr. Pharm. Des. 2013, 19, 4140–4151. [Google Scholar] [CrossRef] [PubMed]
  160. Kodroń, A.; Kowalski, K.; Mussulini, B.H.M.; Hazir, C.; Borrero-Landazabal, M.A.; Ngati, S.; Wasilewski, M.; Chacinska, A. Immunoproteasome-specific Subunit Alterations as a Potential Therapeutic Target for Mitochondriopathies. J. Mol. Biol. 2025, 437, 169229. [Google Scholar] [CrossRef]
  161. Tzemach, R.; Gur, C.; Phan, T.S.; David, E.; Zada, M.; Shmueli, M.D.; Mazuz, K.; Sheban, F.; Kurilovich, A.; Ben Yehuda, M.; et al. Dissection of the immune landscape in psoriatic arthritis defines immunoproteasome up-regulation in treatment resistance. Sci. Immunol. 2025, 10, eadu0284. [Google Scholar] [CrossRef] [PubMed]
  162. Basler, M.; Groettrup, M. Recent insights how combined inhibition of immuno/proteasome subunits enables therapeutic efficacy. Genes Immun. 2020, 21, 273–287. [Google Scholar] [CrossRef] [PubMed]
  163. Basler, M.; Lindstrom, M.M.; LaStant, J.J.; Bradshaw, J.M.; Owens, T.D.; Schmidt, C.; Maurits, E.; Tsu, C.; Overkleeft, H.S.; Kirk, C.J.; et al. Co-inhibition of immunoproteasome subunits LMP2 and LMP7 is required to block autoimmunity. Embo Rep. 2018, 19, e46512. [Google Scholar] [CrossRef] [PubMed]
  164. Johnson, H.W.B.; Lowe, E.; Anderl, J.L.; Fan, A.; Muchamuel, T.; Bowers, S.; Moebius, D.C.; Kirk, C.; McMinn, D.L. Required Immunoproteasome Subunit Inhibition Profile for Anti-Inflammatory Efficacy and Clinical Candidate KZR-616 ((2S,3R)-N-((S)-3-(Cyclopent-1-en-1-yl)-1-((R)-2-methyloxiran-2-yl)-1-oxopropan-2-yl)-3-hydroxy-3-(4-methoxyphenyl)-2-((S)-2-(2-morpholinoacetamido)propanamido)propenamide). J. Med. Chem. 2018, 61, 11127–11143. [Google Scholar] [CrossRef] [PubMed]
  165. Besse, A.; Besse, L.; Kraus, M.; Mendez-Lopez, M.; Bader, J.; Xin, B.T.; de Bruin, G.; Maurits, E.; Overkleeft, H.S.; Driessen, C. Proteasome Inhibition in Multiple Myeloma: Head-to-Head Comparison of Currently Available Proteasome Inhibitors. Cell Chem. Biol. 2019, 26, 340–351.e3. [Google Scholar] [CrossRef]
Figure 1. Simplified schematic of the immunoproteasome structure.
Figure 1. Simplified schematic of the immunoproteasome structure.
Cells 14 01586 g001
Figure 2. Schematic representation of the roles of immunoproteasomes (IMPs) in skeletal muscle pathologies. IMPs play dual roles in skeletal muscle pathologies, supporting repair and homeostasis on one side but driving inflammation and degeneration on the other, with therapeutic strategies aiming to balance these opposing effects.
Figure 2. Schematic representation of the roles of immunoproteasomes (IMPs) in skeletal muscle pathologies. IMPs play dual roles in skeletal muscle pathologies, supporting repair and homeostasis on one side but driving inflammation and degeneration on the other, with therapeutic strategies aiming to balance these opposing effects.
Cells 14 01586 g002
Table 1. Effects of immunoproteasome inhibitors in inflammation models.
Table 1. Effects of immunoproteasome inhibitors in inflammation models.
Name and TargetSelectivityModelEffects and MechanismsRefs.
ONX-0914, LMP2, and LMP7LMP7 subunit (20- to 40-fold more selective than β5c or LMP2)Murine PBMC culture; murine collagen-induced arthritis (CIA)Reduces the production of IL-23 by activated monocytes and IFNγ and IL-2 by T cells. Attenuates the severity of arthritis, reduces cellular infiltration, IL-1β, IL-6, and TNFα production, and autoantibody levels.[78]
ONX-0914 Murine lupus modelPrevents nephritis progression, decreases serum autoantibody levels, and reduces IFNα production of TLR-activated plasmacytoid dendritic cells.[101]
ONX-0914 Murine models of psoriasis, spontaneously developing and imiquimod-inducedReduces skin thickness, inflammation scores, and pathological lesions, normalizes the expression of several pro-inflammatory genes in the ear and significantly reduced the inflammatory infiltrate, accompanied by a significant alteration in the αβ+ and γδ+ T cell subsets.[102]
ONX-0914 Murine models of atherosclerosisReduces atherosclerosis, dendritic cell, and macrophage levels and their activation and the levels of antigen-experienced T cells and Th1 cells. Reduces accumulation of neutrophils and macrophages in white adipose tissue, intestinal triglyceride uptake and gastric emptying, and improves markers of metabolic syndrome.[103]
ONX-0914 Human skeletal muscle myoblastsRecovers reduced cell viability after LMP7 overexpression.[104]
ONX-0914 PBMCs of immune thrombocytopenia (ITP) patients; ITP murine modelIncreases the number of platelets, decreases the expression of FcγRI in ITP mice and decreases that of FcγRIII in ITP patients, inhibits the activation of CD4+ T cells, and affects the differentiation of Th1 cells in patients with ITP.[105]
ONX-0914LU-102, inhibitor of β2 (MECL-1)Human LN229, GBM8401, and U87MG glioblastoma cells; orthotopic mouse glioblastoma modelInduces cell cycle arrest, apoptosis, and autophagy; reduced BCL-2 expression of ONX-0914 also induced in glioblastoma cells. In vivo, TMZ plus ONX-0914 reduced tumor progression better than the control or TMZ alone.[106]
ONX-0914Castration-resistant prostate cancer (CRPC) tumor graft modelSuppresses the “tumor-elicited” Th17-type inflammatory response, angiogenesis, and epithelial–mesenchymal transition via inactivation of COX-2/VEGF-A signaling and β-catenin/Snail signaling.[107]
ONX-0914 and LU-102Human multiple myeloma (MM) cell linesONX-914 induces MM cell cytotoxicity, which is enhanced by IFNγ. LU-102, and dramatically sensitizes MM cells ONX-0914. ONX-0914 synergizes with all FDA-approved proteasome inhibitors in MM in vitro and in vivo.[108]
YU102 and LMP7Murine model of LPS-induced neuroinflammationAttenuates disease progression, reduces the number of reactive astrocytes and microglia, and suppresses the secretion of IL-1α, IL-1β, and CCL12 from microglial cells.[109]
KZR-616,
LMP2, and
LMP7
LMP7 and LMP2 subunits (18- and 81-fold more selective than β5c and β1c)Murine lupus model, healthy volunteersCompletely resolves proteinuria mediated by alterations in T and B cell activation, including reduced numbers of short- and long-lived plasma cells in mice. Selectively inhibits immunoproteasomes and blocks cytokine production following ex vivo stimulation.[81]
KZR-616 Murine model of C-protein-induced myositisSimilarly to ONX 0914, it prevents loss of grip strength, reduces leukocyte infiltration of the muscle, and prevents increased serum creatine kinase levels.[30]
PKS3053 and LMP7LMP7 subunit (5600- and 13,600-fold more selective than β5c)Human PBMCs, murine model of cutaneous lupus erythematosusReduces TLR-dependent activation of plasmacytoid dendritic cells and decreases their maturation, IFNα response, and T cell proliferation. Decreases inflammation, cellular infiltration, and skin damage.[110]
M3258 and LMP7LMP7 subunit (>500-fold more selective than β5c)Human breast cancer (BC) samples and cell lines, murine BC modelReduces viability and induced cell apoptosis in vitro, reduces tumor growth and the tumor abundance of M2 macrophages, activates tumor-infiltrating CD8+ T cells, and suppresses the expression of specific inflammatory pathway gene signatures in immune cells.[111]
DPLG3 and LMP7LMP7 subunit (7200-fold more selective than β5c)Murine dextran sulphate (DSS)-induced colitisAttenuates disease progression and decreases the production of IL-6, IL-1β, IFNγ, and TNFα and the influx of effector T cells and macrophages in colon tissues while increasing the number of Tregs; reduces the expression of NF-κB p50 and p65.[112]
Abbreviations: CCL12, chemokine (C-C motif) ligand 12; PBMC, peripheral blood mononuclear cells; TLR, toll-like receptor; Tregs, T-regulatory cells.
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

Kalinkovich, A.; Livshits, G. Immunoproteasomes in Skeletal Muscle Pathologies: Emerging Roles, Conflicting Evidence, and Future Directions. Cells 2025, 14, 1586. https://doi.org/10.3390/cells14201586

AMA Style

Kalinkovich A, Livshits G. Immunoproteasomes in Skeletal Muscle Pathologies: Emerging Roles, Conflicting Evidence, and Future Directions. Cells. 2025; 14(20):1586. https://doi.org/10.3390/cells14201586

Chicago/Turabian Style

Kalinkovich, Alexander, and Gregory Livshits. 2025. "Immunoproteasomes in Skeletal Muscle Pathologies: Emerging Roles, Conflicting Evidence, and Future Directions" Cells 14, no. 20: 1586. https://doi.org/10.3390/cells14201586

APA Style

Kalinkovich, A., & Livshits, G. (2025). Immunoproteasomes in Skeletal Muscle Pathologies: Emerging Roles, Conflicting Evidence, and Future Directions. Cells, 14(20), 1586. https://doi.org/10.3390/cells14201586

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