Next Article in Journal
The Effect of Physical Activity Interventions on Executive Function in Overweight and Obese Adults: A Systematic Review
Previous Article in Journal
The Role of Regulatory B Lymphocytes in Allergic Diseases
Previous Article in Special Issue
AT2R Activation Improves Wound Healing in a Preclinical Mouse Model
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Critical Analysis of Cytoplasmic Progression of Inflammatory Signaling Suggests Potential Pharmacologic Targets for Wound Healing and Fibrotic Disorders

by
Michael L. Samulevich
1,†,
Liam E. Carman
1,† and
Brian J. Aneskievich
2,*
1
Graduate Program in Pharmacology & Toxicology, University of Connecticut, Storrs, CT 06269-3092, USA
2
Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT 06269-3092, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomedicines 2024, 12(12), 2723; https://doi.org/10.3390/biomedicines12122723
Submission received: 30 October 2024 / Revised: 22 November 2024 / Accepted: 26 November 2024 / Published: 28 November 2024
(This article belongs to the Special Issue Skin Fibrosis and Cutaneous Wound Healing)

Abstract

:
Successful skin wound healing is dependent on an interplay between epidermal keratinocytes and dermal fibroblasts as they react to local extracellular factors (DAMPs, PAMPs, cytokines, etc.) surveyed from that environment by numerous membrane receptors (e.g., TLRs, cytokine receptors, etc.). In turn, those receptors are the start of a cytoplasmic signaling pathway where balance is key to effective healing and, as needed, cell and matrix regeneration. When directed through NF-κB, these signaling routes lead to transient responses to the benefit of initiating immune cell recruitment, cell replication, local chemokine and cytokine production, and matrix protein synthesis. The converse can also occur, where ongoing canonical NF-κB activation leads to chronic, hyper-responsive states. Here, we assess three key players, TAK1, TNFAIP3, and TNIP1, in cytoplasmic regulation of NF-κB activation, which, because of their distinctive and yet inter-related functions, either promote or limit that activation. Their balanced function is integral to successful wound healing, given their significant control over the expression of inflammation-, fibrosis-, and matrix remodeling-associated genes. Intriguingly, these three proteins have also been emphasized in dysregulated NF-κB signaling central to systemic sclerosis (SSc). Notably, diffuse SSc shares some tissue features similar to an excessive inflammatory/fibrotic wound response without eventual resolution. Taking a cue from certain instances of aberrant wound healing and SSc having some shared aspects, e.g., chronic inflammation and fibrosis, this review looks for the first time, to our knowledge, at what those pathologies might have in common regarding the cytoplasmic progression of NF-κB-mediated signaling. Additionally, while TAK1, TNFAIP3, and TNIP1 are often investigated and reported on individually, we propose them here as three proteins whose consequences of function are very highly interconnected at the signaling focus of NF-κB. We thus highlight the emerging promise for the eventual clinical benefit derived from an improved understanding of these integral signal progression modulators. Depending on the protein, its indirect or direct pharmacological regulation has been reported. Current findings support further intensive studies of these points in NF-κB regulation both for their basic function in healthy cells as well as with the goal of targeting them for translational benefit in multiple cutaneous wound healing situations, whether stemming from acute injury or a dysregulated inflammatory/fibrotic response.

1. Introduction

Wound healing is a vital and complex physiological response to injury employed to restore the integrity of damaged tissues across diverse organ systems [1]. Because of their accessible position for observation, experimental investigations, and clinical intervention, the wound healing process involving epithelial and underlying connective tissues is of great interest [2]. Epithelial tissue includes both the keratinized epithelium of the skin and the non-keratinized epithelium of moist regions, e.g., the oral cavity and esophagus. Connective tissue, in this respect, mostly involves fibrous compartments (fibroblasts producing collagen and other extracellular matrix (ECM) components), which, for the skin, are recognized in papillary and reticular layers. Skin keratinocytes and fibroblasts are acknowledged as individually important in situ for tissue and in cell culture model systems of normal and aberrant wound healing and fibrotic responses. Nevertheless, of increasingly recognized significance is their cell–cell cross-communication, resulting in their both secreting and responding to numerous cytokines, chemokines, and matrix proteins [3,4,5,6,7,8,9]. These numerous and varied signaling molecules then contribute to differing phases of the wound healing process. Given the likely relationship of aberrant wound healing to certain skin fibrotic disorders (as we will expand in Section 2.1), we will emphasize cutaneous tissue from here forward.
The canonical wound healing process comprises hemostatic, inflammatory, proliferative, and maturation/remodeling phases [2,10]. Hemostasis, or cessation of bleeding, is achieved via primary and secondary hemostatic processes. Primary hemostasis refers to the initial aggregation of platelets at the site of injury to form a barrier protecting the exposed endothelium in conjunction with brief arterial vasoconstriction [11]. Secondary hemostasis sees activation of both the longer intrinsic and shorter extrinsic clotting pathways, which converge with the proteolytic activation of clotting factor X into factor Xa [12]. The cascade of conversions subsequent to the activation of factor Xa is common to both the intrinsic and extrinsic pathways. These ultimately result in the activation of fibrinogen into fibrin as well as factors that bind the fibrin strands into a fibrin mesh; this mesh stabilizes the platelet plug formed during primary hemostasis.
The inflammatory phase begins with the completion of the hemostatic stage, as thrombocytes involved in the process of hemostasis, along with resident skin cells such as keratinocytes, macrophages, dendritic cells, and mast cells are exposed to both damage- and pattern-associated molecular patterns (DAMPs and PAMPs, respectively) [13]. Pattern recognition receptors (PRRs) on the surface and endosomal membranes of these cells, e.g., Toll-like receptors (TLRs), recognize these DAMPs and PAMPs, triggering an intracellular inflammatory signaling cascade (Figure 1 and our review [14]). This leads to the release of cytokines and chemokines, which trigger local vasodilation and the chemotaxis of additional immune cells, e.g., neutrophils and monocytes, along with the continued inflammatory signaling of resident cells. Negative regulatory processes within cells prevent runaway inflammation, including the interruption of Transforming Growth Factor-β (TGF-β) Activated Kinase 1 (TAK1)’s interaction with polyubiquitin by the ubiquitin editing enzyme Tumor Necrosis Factor-α-Interacting Protein 3 (TNFAIP3, alias A20), whose function is facilitated by TNFAIP3-Interacting Protein 1 (TNIP1, alias ABIN1) (Figure 1). These proteins play a pivotal role in the regulating positive (TAK1) or negative (TNFAIP3, TNIP1) progression of NF-κB signaling. Nevertheless, they have scarcely been considered as therapeutic targets. Further, we suggest here that because of their inter-related but not redundant functions centered on NF-κB function, an integrated review of their role in that pathway, including the current, emerging, or potential opportunities to pharmacologically regulate them, could establish them as new targets for clinical modulation. As such, the cell physiological role and consequences of insufficiency and/or dysfunction of each of these proteins will be discussed in later sections.
The proliferative phase for cutaneous healing begins within a day of injury with the migration and proliferation of wound edge keratinocytes to cover the wound surface in a process called re-epithelialization. This expansion is driven by a lack of contact with the cells lost in the wound site, signaling cytoskeletal reorganization [15]. When the wound edge keratinocytes contact other keratinocytes, their migration ends, and they begin secreting ECM proteins contributing to the basement membrane [13]. It is also in this proliferative phase that fibroblasts migrate to the platelet plug and secrete proteases to remove it while also secreting components of the ECM to form granulation tissue. Finally, the maturation/remodeling phase sees the differentiation of fibroblasts in granulation tissue into myofibroblasts, which produce α-smooth muscle actin (α-SMA) to contract the wound [16].
Although brief, this overview of the classical four stages typical for effective cutaneous wound healing has nevertheless highlighted sufficient and progressive initiation and resolution of each phase [17]. As such, deficiencies or excesses at any stage can and do lead to chronic lesions of incomplete closure or hyper-responsiveness. The first of these two outcomes has been comprehensively reviewed [18,19,20]. Looking forward to the specifics presented below, we will survey excessive responses, especially those where NF-κB has been a focus of investigation. At the cellular and tissue level, this focus will lead us to unresolved, chronic inflammation, which so often accompanies transient dysregulated wound healing (see Section 2.1) and chronic fibrotic disorders (see Section 2.3), in particular for our consideration, systemic sclerosis (SSc, scleroderma).

2. Aberrant Wound Healing

2.1. Wound Healing Gone Wrong

Excessive inflammatory and fibrotic gene expression can underlie unsuccessful skin wound healing, contributing to chronic wounds. These endpoints may arise from diverse conditions of poor glucose regulation associated with diabetes and foot ulcers, surface compression leading to pressure ulcers on bony prominences, and chronic venous insufficiency predisposing to stasis ulcers on the lower extremities [17,21,22,23,24]. With regard to the inflammatory phase, there is something of a “golden mean” in terms of reaching regulatory homeostasis; too little or too much inflammatory signaling results in chronic inflammation, with runaway inflammatory signaling potentially leading to fibrosis. Indeed, Longaker and colleagues [25] recently remarked on wounds either “under-healing” (i.e., showing significant delays or complete failure to resolve) or “over-healing” (marked by scarring and/or fibrosis).
The latter two stages of wound healing, proliferation and remodeling, are also of particular interest in the investigation of chronic wounds as these lesions are typified by failure to progress to these stages [26]. Nevertheless, even successfully closed wounds regularly only reach ~80% restored tensile strength and are almost always marked by scarring [27,28] with parallel bundles of collagen rather than a multidirectional matrix of proteins. In some cases, these scars can be “hypertrophic”, resulting from an imbalance of ECM deposition and degradation in the remodeling phase [28,29]. These share some similarities in appearance to “keloids” but are distinct in terms of boundary and morphology, which are less defined for keloids and not limited to the site of initial injury [30].
The major cell type at play in driving fibrotic outcomes is myofibroblasts [31]. Myofibroblasts are phenotypically fibroblasts that have highly elevated levels of α-SMA chiefly, although other contractile proteins like myosin heavy chain isoforms can be involved [16,32]. These cells may be the products of differentiated fibroblasts but have also been shown to originate from epithelial and endothelial cells, fibrocytes, and smooth muscle cells [33,34]. Their major functions are producing collagens type I and II and providing contractile force; while doing so, they release cytokines such as TGF-β and EGF, amongst others [35,36]. When wound resolution progresses healthily, myofibroblasts undergo apoptosis, and as such, resistance to apoptosis by myofibroblasts is associated with fibrotic disease [37].
Though myofibroblasts are seen as the principal drivers of wound resolution and fibrosis, the independent and cooperative roles keratinocytes and fibroblasts play should not be overlooked [5,16,31]. Keratinocytes, the major constituent cell of the epidermis, serve as the first line of physical defense against pathogens but also have diverse roles in wound healing. These include innate immune functions through surveying their local environment via PRRs, e.g., TLRs [38,39,40] and secretion of cytokines, chemokines, and matrix metalloproteinases (MMPs), all of which are endpoints of keratinocyte inflammatory signaling [38,40,41,42]. Cytoplasmic signaling pathways to get to these endpoints thus offer potential entry points for pharmacologic regulation, especially to address “under-” and “over-healing” in chronic wounds or cutaneous fibrotic disorders such as scleroderma.

2.2. Systemic Sclerosis—Scleroderma

Broadly defined, there are two major categorizations of tissue hardening and fibrosis referred to as scleroderma. The first mostly affects cutaneous tissues, i.e., localized scleroderma. The second is excessive and hardened matrix proteins subtending numerous internal epithelial layers (e.g., skin, gastrointestinal tract, and lungs) but also blood vessel walls (i.e., systemic sclerosis [43]). Given cutaneous involvement in both groups, we inclusively use the term systemic sclerosis (SSc).
SSc is a poorly understood, chronic pathology marked by inflammation and immune system activation, dysfunction in microvascular cells and neighboring fibroblasts, and excess collagen deposition (fibrosis) in the dermis and internal organs [9,44,45,46,47,48]. Within this overall description, there is significant patient-to-patient diversity, important not only for definitive clinical classification but also for patient prognoses. Connective tissue fibrosis, particularly within the dermis, leading to skin hardening, is prototypical for the disease. The etiology, clinical pathology, and cell biology of SSc suggest a “perfect storm” of pre-disposing genetic factors, environmental stressors, and multi-level, multicell signaling interactions coming together to trigger inflammatory signals, microvascular damage, and fibrotic responses where wound healing resolution fails to occur [9,44,45,46,47,48,49]. Reminiscent of the “over-healing” suggested for excessive matrix deposition of terminating regular wound healing, SSc has also been considered a fibrotic stage that fails to resolve [4,50,51], possibly in part driven by ongoing TLR stimulation from otherwise unremarkable levels of DAMPs and PAMPs.
Advances have been made in understanding cell–cell interactions in producing and responding to inflammatory and fibrotic signals in SSc as key to tissue decline in multiple and diverse organs [45,50,52]. Nevertheless, there are significant treatment shortcomings, too often with limited initial or ongoing benefit to individuals living with SSc [53,54,55]. In brief, current treatment approaches for SSc slow the progression of this multifaceted disorder but do little to halt it [47,56,57]. These tactics include diverse blood pressure medications to address narrowed or constricted blood vessels associated with skin and lung complications, steroidal and non-steroidal anti-inflammatories, immune system suppressants, and medications for gastrointestinal tract symptoms [47,57,58]. Such drugs can have significant negative side effects [59,60,61]. Some, like cyclo-phosphamide, raise concerns of carcinogenesis, limiting their consideration and duration [57]. Pharmacologic and biologic intervention of pathway signaling, e.g., nintedanib, a kinase inhibitor for VEGF, FGF, and PDGF receptors, and tocilizumab, a humanized anti-IL-6 receptor monoclonal antibody, hold some promise for certain aspects of SSc. Nevertheless, each of these two treatments is again associated with adverse reactions, such as gastrointestinal side effects and serious infections, respectively [57,62,63].

2.3. Care Cost Impacts of Deficient Wound Healing and Fibrotic Disorders

In addition to the cell biology and medical science challenges in achieving appropriate wound resolution, there are the collateral difficulties of significant health care costs for acute wounds, especially chronic ulcerative lesions and inflammatory/fibrosing disorders such as SSc. While valuable for some comprehensive evaluation of care costs, there is limited availability of broad-based retrospective studies and/or they may be dated or difficult to use for estimation of current costs [64,65,66]. Spending from federal health insurance alone for wound care was estimated at USD 28 billion in a 2018 report and possibly up to three times this depending on diagnosis code criteria [67]. Costs are further exacerbated by infection, patient age, and comorbidities [65].
Comprehensive and directly comparable studies of costs for fibrosing disorders are limited [68,69]. Importantly, by the nature of the pathology, care costs can extend beyond the skin because of frequent internal organ dysfunction such as lung fibrosis, pulmonary arterial hypertension, and heart fibrosis. Significantly higher annual medical costs (reflecting “healthcare resource utilization”) are reported for people living with SSc than those not experiencing it [70,71,72]. In 2023, individual patient costs were reported to reach USD 30,000 [73]. The personal quality of life reductions and financial costs have spurred the identification and development of new therapeutics capitalizing on distinct targets built into cell signaling pathways inherent in regulating SSc inflammatory and fibrotic responses. Therapeutic success in the SSc arena may have benefits translatable to over-healing, scarring and keloids, and, in general, wound healing.

2.4. Consideration of Novel Therapeutics

There is an omnipresent call for novel drug development in targeting pathways [74,75,76,77], which would have therapeutic relevance to aspects of the excess inflammation and matrix deposition shared in both aberrant wound healing and SSc. Longaker and colleagues recently noted wound healing is “the largest medical market without an existing small molecule/drug treatment” [78]. These patient care realities highlight the need for the basic science investigation of cell signaling pathways relevant to effective wound healing that would lead to therapeutics to complement or synergize with current approaches. There has been some advancement in this regard. For instance, the potential for wound healing benefit via the regulation of proteins involved in local signaling was demonstrated by inhibiting Yes-associated protein with verteporfin, which in turn prevented Engrailed-1 expression [79]. This leads to a reduction in the usual fibrosis associated with scarring during wound healing, which is causative of reduced tissue flexibility and mechanical strength [80,81]. The possibility of reducing fibrosis brought about by verteporfin investigations highlights what opportunities may arise from more repurposing of approved drugs and natural products [82,83]. Verteporfin is mostly known for its photosensitizing effect, but it is increasingly being recognized for affecting cytoplasmic signaling pathways [84,85].
Beyond small molecule drugs that themselves might serve to facilitate any stages of wound healing is a prior and building interest [83] in the delivery of those compounds in formulations including but not limited to planar patches, moldable and injectable hydrogels, and nanoparticles that would provide for their local delivery in higher concentrations and rates of release from the drug-containing matrix. These delivery strategies might make possible the effective use of repurposed drugs not otherwise practical because of solubility or adverse events from systemic administration. Lastly, it is also important to note that conceptualizing the source of signaling initiators should not just be limited to soluble factors and cell or infecting bacteria debris. Wound physical tension or pressure, i.e., mechanotransduction, yields biochemical mediators central to the inflammation upstream of fibrosis [86].
Small molecule approaches are also being examined to advance SSc treatment options. Preclinical and trial results, including work with JAK inhibitors ([87,88] for example) and cannabinoid type 2 receptor agonists [89,90,91,92,93], highlight opportunities for SSc therapeutics development well outside current protocols, e.g., steroidal and non-steroidal anti-inflammatories and broad-based chemical and biologic immune system suppressants. For instance, lenabasum, an oral non-immuno-suppressive cannabinoid type 2 receptor (CB2) agonist, had phase 3 trial results recently reported [70]. Although with strong support from preclinical investigations demonstrating a reduction in inflammation and associated fibrosis markers [71] along with being well tolerated [72,94], the study does not show efficacy in diffuse cutaneous systemic sclerosis (dcSSc) [70]. Results may have been obscured by participants’ ongoing treatment regimens, including immuno-suppressive therapies. Separately, there are 2024 preclinical results [73] with CB2 agonist HU-308 diminishing skin and lung fibrosis in a bleomycin fibrosis mouse model.

2.5. Cytoplasmic Regulation of Post-Receptor Signaling May Provide New Opportunities

Given the as-of-yet unmet need for novel anti-inflammatory and antifibrotic therapeutics, additional intracellular pathways post the activation of membrane receptors are also being considered. A prime target is the NF-κB inflammatory pathway downstream of TLR, TGF-βR, and TNF-R, among others as it has been linked to wound healing inflammation and fibrosis as well as to SSc [95,96,97].
As outlined in Figure 1, the central hub of NF-κB signaling is the IκB kinase (IKK) complex (proteins in green), consisting of IKK-α and -β, the effector serine–threonine kinases, and their regulatory subunit IKK-γ, known more commonly as NF-κB essential modulator (NEMO) [98]. This complex is common to all pathways of upstream stimulators of NF-κB signaling, including, but not limited to, the TLR, TNFR, and TGFBR pathways (shown in Figure 1) [98,99]. Upon phosphorylation of the IKK-α and -β subunits, this complex can then phosphorylate downstream machinery such as inhibitor of κB (IκB) family proteins such as IκBα (shown in pink). Subsequently, these proteins are ubiquitinated by E3 ubiquitin ligase complex SCFβTrCP and degraded by the proteasome [100]. This then allows the nuclear translocation of NF-κB homo- and heterodimers (which are sequestered in the cytoplasm in resting cells), allowing them to act as transcription factors for a plethora of pro-inflammatory genes [101]. The NF-κB family consists of five different monomers: NFKB1 (p50/p105), NFKB2 (p52/p100), RelA (p65), c-Rel, and RelB [96]; the p50 and p52 subunits must also be cleaved via the proteasome, constitutively or upon stimulus, from their p105 and p100 precursors, respectively, in order to be active [102,103,104]. The p50/p65 dimer is shown in Figure 1 and is commonly cited as the most abundant, although this can vary based on cellular context [105].
Myriad genes regulated by NF-κB transcription factors downstream of surface receptor signaling experience increased expression during the wound healing process [101,106,107]. For example, CCR2 is a chemokine receptor with activity critical to early vascular sprouting. It has been demonstrated to both have increased expression in the tissue of patients during wound healing post burn injury and result in deficient wound healing with its knockout in murine studies [108,109,110]. The SERPINE1 gene has also been shown to be highly upregulated following suction-blister wound infliction [111]. PAI-1, the protein encoded by the SERPINE1 gene, is instrumental in re-epithelialization through its roles in both cell movement and tissue remodeling [112,113]. Unsurprisingly, the inflammatory cytokine TNF-α has been shown to be rapidly upregulated in wounded tissues and directly impacts wound healing through its acceleration of wound re-epithelialization and neovascularization [114,115]. Since runaway signaling in the inflammatory phase of wound healing has been linked to chronic and fibrotic wounds, there has been an investigation into the inhibition of TNF-α as a therapeutic avenue [116]. NF-κB activation is, thus, a centralization point of diverse extracellular signals leading to activation of inflammatory and fibrotic signals. Below, we survey TAK1, TNFAIP3, and TNIP1 as cytoplasmic proteins overseeing that activation with an eye toward the pharmacologic potential of this pathway (Figure 1). Although beyond the immediate scope of this review, we nevertheless stress the importance of a holistic appreciation of these three proteins in other signaling pathways, e.g., those mediated by MAPK or those leading to necroptosis or mitophagy (for review, see [117,118,119,120]).

3. Association of NF-κB Signal Repressing Proteins with SSc

3.1. TAK1

TGF-β-activated kinase 1 (TAK1, alias MAP3K7) was first identified in mice as a unique member of the mitogen-activated protein kinase (MAPK) family, which participates in the mediation of TGF-β superfamily signaling [121]. The human homolog of TAK1 was later shown to activate kinases downstream in multiple modes of inflammatory signaling, such as the IL-1β, NF-κB, and multiple MAPK pathways [122,123]. Comprehensively, TAK1 is now understood to be a central component in both cell death and inflammatory signaling in a multitude of cell types and disease states [117,124,125].
In signaling paradigms such as the IL-1β, TLR, TNF-R, and non-canonical TGF-β pathways, receptor activation leads to the recruitment of E3 ubiquitin ligases such as TRAF6, which form polyubiquitin chains [126,127]. The TAK1 binding protein family (TAB), namely members TAB2 and 3, bind the ubiquitin chains and recruit TAK1 to phosphorylate and activate downstream components, such as the IKK complex in NF-κB signaling and MAPKs, to progress signaling (Figure 1) [127,128,129]. While inflammatory signaling may carry a negative connotation, components of such signaling, including TAK1, play important roles in normal cell processes such as cell proliferation and survival, innate immunity in pathogen defense, and myocardial function [123,130,131]. This is demonstrated especially in wound healing, wherein TAK1 in dermal fibroblasts supports their proliferation and survival, the deposition of ECM, and ultimately the contraction of healing wounds [132].
The contribution to fibroblast function that TAK1 possesses is, of course, dependent on a delicate balance between sufficient and excessive signaling. Dysregulated wound healing, characterized by abnormalities in the newly formed fibronectin matrix (namely the presence of fibronectin EDA and unfolded type III fibronectin, recognized as DAMPs) yields excessive inflammatory signaling in fibroblasts through the TLR4/TAK1 axis [133]. The effects of fibronectin DAMPs varied by fibroblast origin, with dermal fibroblasts responding to FnEDA with the induction of the IL-1β and CCL5 genes [133], the latter protein having been implicated in the progression of scleroderma [3].
TAK1 mediates both TLR and TGF-β signaling (Figure 1) making it an enticing target for multiple modes of tissue fibrosis [125,134]. It comes as no surprise that experimental small molecule inhibitors of TAK1, such as HS-276 [134,135], have shown promise by preventing TGF-β-stimulated collagen synthesis and myofibroblast differentiation in control skin fibroblasts as well as mitigating the profibrotic phenotype in SSc fibroblasts and bleomycin mouse models (see Asano et al. [136] for further discussion of TAK1 pharmacologic inhibition). Interestingly, the inhibition of TAK1 with HS-276 concomitant with TGF-β exposure in fibroblasts resulted in the upregulation of some protective anti-inflammatory genes, such as TNFAIP3 [134]. This suggests a synergistic effect of TAK1 inhibition, calming the progression of inflammatory signaling while also enhancing cells’ ability to dampen it. While other inhibitors of TAK1 have been developed, they have not shown the selectivity and bioavailability needed for clinical application [137]. Notably, HS-276 boasts an oral bioavailability of >95% and strong selectivity for TAK-1 over close homologs [134,138]; as such, it recently gained a U.S. FDA Orphan Drug Designation as EYD-001 [139].
TAK1 inhibition may also prove useful for reducing or preventing scar formation given its activity in dermal fibroblasts with the myofibroblast phenotype [140]. Salinomycin and related polyether ionophore antibiotics have been shown to reduce the activation of TAK1 as well as p38 in this signaling cascade [141]. Additionally, TGF-β-stimulated NIH 3T3 mouse fibroblasts showed reduced proliferation and collagen expression when treated with angiotensin-converting enzyme inhibitors (ACEIs) like lisinopril with the concomitant suppression of TAK1 phosphorylation [142]. siRNA knockdown of proteins CPEB1/4 acts similarly in suppressing TAK1 and SMAD signaling and reduces the expression of myofibroblast markers (α-SMA) that contribute to scarring as well as inflammatory markers like TNF-α and IL-6 [143]. Knockdown of both CPEB1 and 4 was accompanied by a significant reduction in phosphorylation of TAK1, p65, and IκBα, further supporting the involvement of the TAK1-NF-κB inflammatory axis in this effect. Interestingly, exosomes from scar fibroblasts have been shown to induce a profibrotic phenotype in healthy skin fibroblasts via the TAK1 and SMAD pathways, suggesting that small numbers of fibrotic cells can influence larger populations at the site of a wound [144].
In sum, TAK1 is an important central component of both TLR and TGF-β signaling, key in both healthy and fibrotic cell signaling. Its pharmacologic inhibition may provide synergistic anti-inflammatory effects in multifactorial events like wound healing and fibrotic diseases like SSc. Having considered TAK1 as the key kinase driving inflammation and fibrosis, it is important to now give attention to proteins central to the repression of inflammatory signaling as yet unexplored pharmacologic targets.

3.2. TNFAIP3

TNF α-induced protein 3 (TNFAIP3, alias A20) was initially discovered due to its expression in response to TNF stimulation of endothelial cells, leading to its description as an inhibitor of TNF-induced apoptosis [145]. Further investigation elucidated this protective function as a result of TNFAIP3 repressing NF-κB inflammatory signaling downstream of various receptor signaling pathways (e.g., TNF-Rs or TLRs) [118,146]. It was through this finding that TNFAIP3 was recognized as a ubiquitin editing enzyme with ligase and deubiquitinase functions [147]. However, the catalytic N-terminal deubiquitinase domain is largely dispensable for the TNFAIP3 reduction in NF-κB-mediated signaling [14,148]. The anti-inflammatory and, thus, cyto-protective abilities of TNFAIP3 seem to stem primarily from its non-catalytic zinc finger (ZnF) 7 and ZnF 4 polyubiquitin (pUb)-binding domains, suggesting that non-enzymatic protein–protein interactions (PPIs) are key. Importantly [14,148], its overall effect may be dependent on cell type, expression levels, and availability of TNFAIP3-partner proteins, e.g., TNIP1 (Figure 1). The latter apparently can act independently of TNFAIP3, but also with it, to bring about a limitation to NF-κB activation [149], leading to the notion that non-enzymatic TNIP1 serves as a scaffold to facilitate TNFAIP3’s pUb interaction [119].
TNFAIP3 is known to stunt IKK activation by the TAK1:TAB2/3 heterodimer through its recognition of M1-linked (linear) pUb chains via its ZnF7 domain [150]; this interaction (Figure 1) is thought to be strengthened via TNIP1 interaction, as TNIP1 also preferentially recognizes linear pUb chains via its UBAN domain [151,152]. TNFAIP3’s prevention of IKK complex phosphorylation prevents subsequent activation of the NF-κB complex for nuclear localization, thus preventing further NF-κB-mediated inflammatory signaling [101].
The genes whose transcription is upregulated by NF-κB signaling are cell type-dependent, with the pro-inflammatory signaling seen in keratinocytes contributing to the inflammatory phase of wound healing and fibroblasts seeing an increase in not only pro-inflammatory gene expression but also profibrotic gene expression [153,154]. Thus, TNFAIP3-mediated repression of NF-κB signaling is a crucial braking mechanism for multiple phases in overactive wound healing [155].
The need for the crucial negative regulatory capacity of TNFAIP3 in typical wound healing is echoed in SSc as a hyper-inflammatory and fibrotic disorder. Perhaps most telling are the significantly reduced levels of TNFAIP3 mRNA in the transcriptome of SSc skin biopsies [156]. Additionally, making it something of an innate trait rather than some response to the tissue environment, TNFAIP3 expression is decreased in explanted SSc fibroblasts when controlled against healthy explanted fibroblasts [156]. Less immediately obvious but not necessarily less important are TNFAIP3 links to SSc by various genome-wide association studies (GWASs) and other gene polymorphism reports [157,158,159,160,161,162]. In a specific example, the SNP rs58905141 of TNFAIP3 was reported as linked with the expression of MMPs in fibroblasts challenged with silica particles, an environmental factor linked with the development of SSc [163].
Some pharmacological regulation of TNFAIP3 levels, although outside the arena of wound healing, has been reported due to its inflammation-limiting effect in diverse cells and tissues. This appears to be through increasing its expression level, rather than outright small molecule binding to its protein and facilitating its function. For instance, ikarugamycin and quercetin [164] and gibberellic acid [165] induce the expression of TNFAIP3 in culture models employing bronchial epithelial cell lines. This suggests the compounds’ relevance to minimizing the throughput of incoming inflammatory signals associated with cystic fibrosis. Highlighting the need to look beyond typical dermal wound healing reports, Sun and colleagues recently reported [166] on increased TNFAIP3 expression in prostate cancer cells as a consequence of their exposure to the pyrimidine diamine compound ZY-444, also known as an inhibitor of pyruvate carboxylase.
Reduction in NF-κB activation by increased TNFAIP3 expression was also central to inflammatory and fibrotic signal reduction after the stimulation of the adiponectin receptor in dermal fibroblasts. This was achieved initially with the 244-amino acid adipokine hormone and later with a small molecule ligand that binds to and activates the receptor [156,167,168]. The latter is advantageous to translational development for aberrant wound healing or SSc treatments because it would be orally available without the encumbrances of the active agent being a relatively large protein. These and other TNFAIP3 findings above do strengthen its positioning as a wound healing/fibrosis target, especially in the context that enhancing its protein levels for its deubiquitinase activity and/or PPIs may bring about the desired effect. One such PPI is with its partner protein TNIP1, which cooperatively facilitates TNFAIP3’s innate repression of inflammatory signaling [169,170,171].

3.3. TNIP1

The TNIP1 acronym derives from TNF α-induced protein 3-Interacting Protein 1, with TNFAIP3 being a TNIP1-binding protein. TNIP1 protein functions with and, importantly, independently of TNFAIP3 to significantly limit the cytoplasmic progression of inflammatory and fibrotic signals downstream of TLR and TNF-R, as we [39,172] and others reported [151,173]. In brief, TNIP1 terminates post-membrane receptor signals (Figure 1) by assembling protein–protein interactions (PPIs) [119,172,174,175]. This inhibition arises from the required TNIP1 recognition of linear pUb on other proteins and its non-compulsory association with partner protein TNFAIP3 for the termination of membrane receptors’ signal cascades, otherwise leading to NF-κB, p38 MAPK, or JNK activation with subsequent inflammatory and fibrotic responses. Physiologically normal levels of TNIP1 protein significantly limit (Figure 1) the cytoplasmic activation of NF-κB downstream of TLR and TNF-R stimulation in diverse cell types [39,119,171,172,176,177,178,179].
Despite the great potential to quell the cytoplasmic progression of inflammatory signaling, there are very limited TNIP1-dedicated wound healing studies to date. We previously reported [172] that TNIP1-deficient keratinocytes are hyper-responsive to the model DAMP / PAMP poly (I:C), with enhanced expression of wound-associated markers (e.g., TGF-β and CCN2). Nevertheless, in a cell wound healing model, there was restricted re-epithelialization and reduced cell viability, potentially due to increased priming of the inflammasome, suggesting the need for TNIP1’s limiting of TLR-initiated signaling early in and throughout the repair process.
In contrast to the paucity of TNIP1 studies in wound healing per se, it has been repeatedly cited [180,181,182,183,184,185,186] (and see our reviews [14,119]) over the last 13 years as one of the highest-scoring SSc risk loci amongst non-HLA genes in multiple populations from numerous GWASs. Importantly, within these studies are TNIP1 risk loci where, when examined, its protein in SSc tissue was significantly reduced. This was paralleled by its mRNA and protein being reduced in cells cultured from SSc donors to about half that compared to control cells [180]. Separate expression array studies [187] support reduced TNIP1 in affected skin compared to patient-uninvolved skin. The findings are consistent with SNPs that may lessen transcription factor binding at the gene promoter or mRNA intron processing efficiency and/or mRNA half-life, leading to decreased mature mRNA and protein [188].
Regarding the TNIP1 functionality of NF-κB inhibition, this predicts hyper-responsiveness to stimuli activating TLR and TNF-R. Indeed, Allanore and colleagues reported [180] reduced TNIP1 expression in fibroblasts cultured from SSc donors, where the cells produced elevated levels of collagen in response to a TNFα challenge. Importantly, for what may be a runaway loss of TNIP1 in instances where there is a genetic disposition to already reduced levels, TNIP1 is degraded, as we [189], and later others [190], showed, as a consequence of inflammation. Notwithstanding the extensively demonstrated repression of cytoplasmic progression of inflammatory signaling and its involvement in fibrotic responses, it is uncertain to date if TNIP1 will be successfully clinically targeted to augment its expression or function. In sum, TAK1, TNFAIP3, and TNIP1 are all involved in NF-κB cytoplasmic signaling in such a way that repression of TAK1 or facilitation of TNFAIP3 and TNIP1 could repress inflammatory signals.

4. Concluding Observations

The importance of integrated, multidisciplinary approaches to advance wound healing research is evidenced by the current, only partially met, need to achieve optimal or complete extents of tissue repair/regeneration in a timely fashion. This is the case for both recovery from acute injury (physical or thermal trauma) as well as those incidents that arise from persistent conditions (e.g., vascular insufficiency, compromised mobility, or diabetes) that lead to chronic lesions (e.g., venous and/or arterial, decubitus, or diabetic foot ulcers) [18,19] or ongoing hyper-inflammatory and hyperfibrotic disorders (e.g., SSc). For wound healing, current approaches have led to better physical and chemical control of infections along with synthetic and biological barriers to protect and better manage tissue voids while supporting tissue regrowth [22,191,192]. Nevertheless, shortcomings remain in our understanding of macro events as well as intracellular signaling both during sufficient wound healing and especially instances of imbalanced healing and fibrotic disorders.
New areas of research are being informed by investigating intracellular signaling pathways responsible for the discrete phases of both normal and dysregulated wound healing. Prominent in this respect are proteins promoting or restricting NF-κB, given its extensive history of research in balancing transient and chronic inflammation as well as beneficial or excessive matrix deposition [193,194,195,196]. In both of these instances, there are emerging ideas that studies of acute and chronic wounds as well as disorders of inflammation and fibrosis, when considered to be aberrant wound healing, can cross-inform each other [4,50,51,197,198]. Recalling the earlier cited comments of Longaker and colleagues [78] on the need for small molecule therapeutics development in this area, we suggest that small molecule inhibitors of TAK1 being investigated for limiting fibrosis in SSc may result in fruitful studies on the regulation of excess matrix deposition in scarring and keloid formation. Complementing this approach, the augmented expression of TNFAIP3 being investigated in multiple avenues (e.g., SSc, cystic fibrosis) could prove to be productive grounds for dedicated wound healing studies capitalizing on the repurposing of approved or pipeline therapeutics. Lastly, interventions to block NF-κB for the expected advantages of supporting later wound healing phases will likely need to be balanced against desirable NF-κB-mediated outcomes, e.g., migration and/or proliferation [195].

5. Prospectus

A new understanding of the dynamics of wound healing can continue to inform therapeutic options for those experiencing chronic inflammatory or fibrotic disease states. Very recent work using specific cell ablation techniques [199] helps to call for a reconsideration of physical events outside the cell at the tissular level of the skin. They detected unexpected dynamic aspects within the classical wound healing phase progression. Their results support envisioning a pre-wound solid-like homeostatic state. Upon wound healing, a transient fluid-like state is brought about by changes in the local signaling environment that influence inflammation, cell migration, and replication. Ultimately, in successful wound healing, there is a return to a solid-like homeostatic state. The challenge remains now to bridge these newly appreciated tissue physical repair dynamics with the specific signaling pathways responsible for them and how to pharmacologically intervene to address signaling defects leading to the under- or over-healing described above.
In addition to reimagining the physical state of cells as above, there are similarly innovative approaches being taken in other arenas to facilitate wound healing. While outside the pharmacologic scope of this review, naturally derived and engineered biomaterials are being investigated for their ultimate effect of limiting NF-κB expression or its activation [200,201]. Going to the source of proteins, gene editing is also being considered for diverse elements of wound healing [202] including those that drive inflammation. Of particular note is an inflammation-induced CRISPR-Cas9 system [203], where the initiation of inflammatory signaling was itself used as a means to negate the expression of MyD88, an upstream signal relay point in eventual NF-κB activation. Parallel to this, non-coding RNAs and their involvement in the regulation of cell membrane receptor-NF-κB signaling axes are also under investigation [204].
An often-overlooked factor involved in the signaling cascades necessary for proper wound healing is polyubiquitin (pUb). We looked intracellularly at three specific proteins, TAK1, TNFAIP3, and TNIP1, for new opportunities in addressing cytoplasmic pathway regulation central to wound healing and fibrotic disorders. Intriguingly, pUb chains, whether part of protein–protein recognition and interaction or for targeting of protein degradation cellular machinery, affect the function of these three proteins. Polyubiquitin reading (recognition), writing (addition), and erasing (removal) have been functionally summed as responsible for the “ubiquitin code” [205,206]. Other proteins responsible for these functions are likely themselves to be yet more intracellular targets for eventual pharmacologic regulation (direct or indirect depending on the reading/writing/erasing action of the involved protein) to better fulfill phases of wound healing. For instance, neuronally expressed developmentally downregulated 4 (Nedd4) is an E3 ligase (writer) central to corneal epithelial wound healing. Although not yet small molecule-targeted, its natural progression of increased expression during epithelial repair suggests its pharmacologically enhanced expression or activity could further its positive effects in that tissue. Additionally, erasers (deubiquitinating enzymes) are being examined in animal cutaneous wound healing models where, at least for ubiquitin C-terminal hydrolase L1 (UCHL1) [207], inhibition by a small molecule promoted wound healing. Finally, the dedicated writing versus erasing functions of individual proteins such as Nedd4 and UCHL1 participating in the ubiquitin code recall that these two functions as well as pUb recognition have been recognized for TNFAIP3 [14]. It may be that those individual roles are more prominent at distinct points of wound healing as far as guiding their small molecule enhancement or repression.
There is a continuously expanding knowledge base of the dynamics of wound healing, especially from key signaling modulators, such as the TAK1, TNFAIP3, and TNIP1 proteins emphasized in this review. Despite their distinct individual abilities, their functions converge on one of the most universal points of inflammatory signaling in successful wound healing, dysregulated recovery, and fibrotic disorders: cytoplasmic activation of NF-κB. In each instance, their simultaneous consideration as offered in this review is likely to inform future therapeutics development, as is emerging with small molecule regulation, directly or indirectly, for at least some of them. Nevertheless, multipronged and integrated investigations of intracellular events like NF-κB activation and gene targeting should be synergized with extracellular events such as the conceptualization of a quasi-fluidic wound tissue and biomaterial effects to the benefit of realizing better clinical control. Merging these individual investigative avenues could provide a more therapeutically effective “superhighway” beyond the current armamentarium for treating wound healing and fibrotic disorders.

Author Contributions

Conceptualization, B.J.A.; investigation, M.L.S., L.E.C. and B.J.A.; writing—original draft preparation, M.L.S., L.E.C. and B.J.A.; writing—review and editing, M.L.S., L.E.C. and B.J.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

Work cited for TNIP1 in the authors’ laboratory was supported in part by a DoD CDMRP grant W81XWH-20-1-0081 (BJA). MLS and LEC received graduate assistantship support from the Department of Pharmaceutical Sciences. We also appreciate the insightful and thoughtful comments of this article’s peer reviewers.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Stroncek, J.D.; Reichert, W.M. Frontiers in Neuroengineering. Overview of Wound Healing in Different Tissue Types. In Indwelling Neural Implants: Strategies for Contending with the In Vivo Environment; Reichert, W.M., Ed.; Taylor & Francis Group, LLC: Boca Raton, FL, USA, 2008. [Google Scholar]
  2. Rodrigues, M.; Kosaric, N.; Bonham, C.A.; Gurtner, G.C. Wound Healing: A Cellular Perspective. Physiol. Rev. 2019, 99, 665–706. [Google Scholar] [CrossRef] [PubMed]
  3. McCoy, S.S.; Reed, T.J.; Berthier, C.C.; Tsou, P.S.; Liu, J.; Gudjonsson, J.E.; Khanna, D.; Kahlenberg, J.M. Scleroderma keratinocytes promote fibroblast activation independent of transforming growth factor beta. Rheumatology 2017, 56, 1970–1981. [Google Scholar] [CrossRef] [PubMed]
  4. Stone, R.C.; Chen, V.; Burgess, J.; Pannu, S.; Tomic-Canic, M. Genomics of Human Fibrotic Diseases: Disordered Wound Healing Response. Int. J. Mol. Sci. 2020, 21, 8590. [Google Scholar] [CrossRef]
  5. Wojtowicz, A.M.; Oliveira, S.; Carlson, M.W.; Zawadzka, A.; Rousseau, C.F.; Baksh, D. The importance of both fibroblasts and keratinocytes in a bilayered living cellular construct used in wound healing. Wound Repair. Regen. 2014, 22, 246–255. [Google Scholar] [CrossRef] [PubMed]
  6. Raziyeva, K.; Kim, Y.; Zharkinbekov, Z.; Kassymbek, K.; Jimi, S.; Saparov, A. Immunology of Acute and Chronic Wound Healing. Biomolecules 2021, 11, 700. [Google Scholar] [CrossRef] [PubMed]
  7. Smith, J.; Rai, V. Novel Factors Regulating Proliferation, Migration, and Differentiation of Fibroblasts, Keratinocytes, and Vascular Smooth Muscle Cells during Wound Healing. Biomedicines 2024, 12, 1939. [Google Scholar] [CrossRef] [PubMed]
  8. Amiri, N.; Golin, A.P.; Jalili, R.B.; Ghahary, A. Roles of cutaneous cell-cell communication in wound healing outcome: An emphasis on keratinocyte-fibroblast crosstalk. Exp. Dermatol. 2022, 31, 475–484. [Google Scholar] [CrossRef]
  9. Truchetet, M.E.; Brembilla, N.C.; Chizzolini, C. Current Concepts on the Pathogenesis of Systemic Sclerosis. Clin. Rev. Allergy Immunol. 2023, 64, 262–283. [Google Scholar] [CrossRef]
  10. Wallace, H.A.; Basehore, B.M.; Zito, P.M. Wound Healing Phases. In StatPearls; StatPearls Publishing LLC: Treasure Island, FL, USA, 2024. [Google Scholar]
  11. Barmore, W.; Bajwa, T.; Burns, B. Biochemistry, Clotting Factors. In StatPearls; StatPearls Publishing LLC: Treasure Island, FL, USA, 2024. [Google Scholar]
  12. Chaudhry, R.; Usama, S.M.; Babiker, H.M. Physiology, Coagulation Pathways. In StatPearls; StatPearls Publishing LLC: Treasure Island, FL, USA, 2024. [Google Scholar]
  13. Landén, N.X.; Li, D.; Ståhle, M. Transition from inflammation to proliferation: A critical step during wound healing. Cell Mol. Life Sci. 2016, 73, 3861–3885. [Google Scholar] [CrossRef]
  14. Carman, L.E.; Samulevich, M.L.; Aneskievich, B.J. Repressive Control of Keratinocyte Cytoplasmic Inflammatory Signaling. Int. J. Mol. Sci. 2023, 24, 1943. [Google Scholar] [CrossRef]
  15. Jacinto, A.; Martinez-Arias, A.; Martin, P. Mechanisms of epithelial fusion and repair. Nat. Cell Biol. 2001, 3, E117–E123. [Google Scholar] [CrossRef] [PubMed]
  16. Wang, K.; Wen, D.; Xu, X.; Zhao, R.; Jiang, F.; Yuan, S.; Zhang, Y.; Gao, Y.; Li, Q. Extracellular matrix stiffness-The central cue for skin fibrosis. Front. Mol. Biosci. 2023, 10, 1132353. [Google Scholar] [CrossRef] [PubMed]
  17. Peña, O.A.; Martin, P. Cellular and molecular mechanisms of skin wound healing. Nat. Rev. Mol. Cell Biol. 2024, 25, 599–616. [Google Scholar] [CrossRef] [PubMed]
  18. Hofmann, A.G.; Deinsberger, J.; Oszwald, A.; Weber, B. The Histopathology of Leg Ulcers. Dermatopathology 2024, 11, 62–78. [Google Scholar] [CrossRef] [PubMed]
  19. Raffetto, J.D.; Ligi, D.; Maniscalco, R.; Khalil, R.A.; Mannello, F. Why Venous Leg Ulcers Have Difficulty Healing: Overview on Pathophysiology, Clinical Consequences, and Treatment. J. Clin. Med. 2020, 10, 29. [Google Scholar] [CrossRef]
  20. Falanga, V.; Isseroff, R.R.; Soulika, A.M.; Romanelli, M.; Margolis, D.; Kapp, S.; Granick, M.; Harding, K. Chronic wounds. Nat. Rev. Dis. Primers 2022, 8, 50. [Google Scholar] [CrossRef]
  21. Lim, J.Z.; Ng, N.S.; Thomas, C. Prevention and treatment of diabetic foot ulcers. J. R. Soc. Med. 2017, 110, 104–109. [Google Scholar] [CrossRef]
  22. Freedman, B.R.; Hwang, C.; Talbot, S.; Hibler, B.; Matoori, S.; Mooney, D.J. Breakthrough treatments for accelerated wound healing. Sci. Adv. 2023, 9, eade7007. [Google Scholar] [CrossRef]
  23. Al-Rikabi, A.H.A.; Tobin, D.J.; Riches-Suman, K.; Thornton, M.J. Dermal fibroblasts cultured from donors with type 2 diabetes mellitus retain an epigenetic memory associated with poor wound healing responses. Sci. Rep. 2021, 11, 1474. [Google Scholar] [CrossRef]
  24. Rai, V.; Moellmer, R.; Agrawal, D.K. The role of CXCL8 in chronic nonhealing diabetic foot ulcers and phenotypic changes in fibroblasts: A molecular perspective. Mol. Biol. Rep. 2022, 49, 1565–1572. [Google Scholar] [CrossRef] [PubMed]
  25. desJardins-Park, H.E.; Gurtner, G.C.; Wan, D.C.; Longaker, M.T. From Chronic Wounds to Scarring: The Growing Health Care Burden of Under- and Over-Healing Wounds. Adv. Wound Care 2022, 11, 496–510. [Google Scholar] [CrossRef] [PubMed]
  26. Zhao, R.; Liang, H.; Clarke, E.; Jackson, C.; Xue, M. Inflammation in Chronic Wounds. Int. J. Mol. Sci. 2016, 17, 2085. [Google Scholar] [CrossRef] [PubMed]
  27. Rivera, A.E.; Spencer, J.M. Clinical aspects of full-thickness wound healing. Clin. Dermatol. 2007, 25, 39–48. [Google Scholar] [CrossRef]
  28. Marshall, C.D.; Hu, M.S.; Leavitt, T.; Barnes, L.A.; Lorenz, H.P.; Longaker, M.T. Cutaneous Scarring: Basic Science, Current Treatments, and Future Directions. Adv. Wound Care 2018, 7, 29–45. [Google Scholar] [CrossRef]
  29. Zhu, Z.; Ding, J.; Tredget, E.E. The molecular basis of hypertrophic scars. Burns Trauma 2016, 4, 2. [Google Scholar] [CrossRef]
  30. Mony, M.P.; Harmon, K.A.; Hess, R.; Dorafshar, A.H.; Shafikhani, S.H. An Updated Review of Hypertrophic Scarring. Cells 2023, 12, 678. [Google Scholar] [CrossRef]
  31. Knoedler, S.; Broichhausen, S.; Guo, R.; Dai, R.; Knoedler, L.; Kauke-Navarro, M.; Diatta, F.; Pomahac, B.; Machens, H.G.; Jiang, D.; et al. Fibroblasts-the cellular choreographers of wound healing. Front. Immunol. 2023, 14, 1233800. [Google Scholar] [CrossRef]
  32. Chiavegato, A.; Bochaton-Piallat, M.L.; D’Amore, E.; Sartore, S.; Gabbiani, G. Expression of myosin heavy chain isoforms in mammary epithelial cells and in myofibroblasts from different fibrotic settings during neoplasia. Virchows Arch. 1995, 426, 77–86. [Google Scholar] [CrossRef] [PubMed]
  33. Hinz, B.; Phan, S.H.; Thannickal, V.J.; Galli, A.; Bochaton-Piallat, M.L.; Gabbiani, G. The myofibroblast: One function, multiple origins. Am. J. Pathol. 2007, 170, 1807–1816. [Google Scholar] [CrossRef]
  34. Schuster, R.; Younesi, F.; Ezzo, M.; Hinz, B. The Role of Myofibroblasts in Physiological and Pathological Tissue Repair. Cold Spring Harb. Perspect. Biol. 2023, 15, a041231. [Google Scholar] [CrossRef] [PubMed]
  35. Tai, Y.; Woods, E.L.; Dally, J.; Kong, D.; Steadman, R.; Moseley, R.; Midgley, A.C. Myofibroblasts: Function, Formation, and Scope of Molecular Therapies for Skin Fibrosis. Biomolecules 2021, 11, 1095. [Google Scholar] [CrossRef] [PubMed]
  36. Younesi, F.S.; Miller, A.E.; Barker, T.H.; Rossi, F.M.V.; Hinz, B. Fibroblast and myofibroblast activation in normal tissue repair and fibrosis. Nat. Rev. Mol. Cell Biol. 2024, 25, 617–638. [Google Scholar] [CrossRef] [PubMed]
  37. Hinz, B.; Lagares, D. Evasion of apoptosis by myofibroblasts: A hallmark of fibrotic diseases. Nat. Rev. Rheumatol. 2020, 16, 11–31. [Google Scholar] [CrossRef] [PubMed]
  38. Piipponen, M.; Li, D.; Landén, N.X. The Immune Functions of Keratinocytes in Skin Wound Healing. Int. J. Mol. Sci. 2020, 21, 8790. [Google Scholar] [CrossRef] [PubMed]
  39. Rudraiah, S.; Shamilov, R.; Aneskievich, B.J. TNIP1 reduction sensitizes keratinocytes to post-receptor signalling following exposure to TLR agonists. Cell. Signal. 2018, 45, 81–92. [Google Scholar] [CrossRef]
  40. Jiang, Y.; Tsoi, L.C.; Billi, A.C.; Ward, N.L.; Harms, P.W.; Zeng, C.; Maverakis, E.; Kahlenberg, J.M.; Gudjonsson, J.E. Cytokinocytes: The diverse contribution of keratinocytes to immune responses in skin. JCI Insight 2020, 5, e142067. [Google Scholar] [CrossRef]
  41. Simmons, J.; Gallo, R.L. The Central Roles of Keratinocytes in Coordinating Skin Immunity. J. Investig. Dermatol. 2024, 144, 2377–2398. [Google Scholar] [CrossRef]
  42. Chieosilapatham, P.; Kiatsurayanon, C.; Umehara, Y.; Trujillo-Paez, J.V.; Peng, G.; Yue, H.; Nguyen, L.T.H.; Niyonsaba, F. Keratinocytes: Innate immune cells in atopic dermatitis. Clin. Exp. Immunol. 2021, 204, 296–309. [Google Scholar] [CrossRef]
  43. Adigun, R.; Goyal, A.; Hariz, A. Systemic Sclerosis (Scleroderma). In StatPearls; StatPearls Publishing LLC: Treasure Island, FL, USA, 2024. [Google Scholar]
  44. Allanore, Y.; Simms, R.; Distler, O.; Trojanowska, M.; Pope, J.; Denton, C.P.; Varga, J. Systemic sclerosis. Nat. Rev. Dis. Primers 2015, 1, 15002. [Google Scholar] [CrossRef]
  45. Bale, S.; Verma, P.; Varga, J.; Bhattacharyya, S. Extracellular Matrix-Derived Damage-Associated Molecular Patterns (DAMP): Implications in Systemic Sclerosis and Fibrosis. J. Investig. Dermatol. 2023, 143, 1877–1885. [Google Scholar] [CrossRef]
  46. Tsou, P.S.; Varga, J.; O’Reilly, S. Advances in epigenetics in systemic sclerosis: Molecular mechanisms and therapeutic potential. Nat. Rev. Rheumatol. 2021, 17, 596–607. [Google Scholar] [CrossRef] [PubMed]
  47. Pope, J.E.; Denton, C.P.; Johnson, S.R.; Fernandez-Codina, A.; Hudson, M.; Nevskaya, T. State-of-the-art evidence in the treatment of systemic sclerosis. Nat. Rev. Rheumatol. 2023, 19, 212–226. [Google Scholar] [CrossRef]
  48. Volkmann, E.R.; Andréasson, K.; Smith, V. Systemic sclerosis. Lancet 2023, 401, 304–318. [Google Scholar] [CrossRef]
  49. Pattanaik, D.; Brown, M.; Postlethwaite, B.C.; Postlethwaite, A.E. Pathogenesis of Systemic Sclerosis. Front. Immunol. 2015, 6, 272. [Google Scholar] [CrossRef]
  50. Bhattacharyya, S.; Varga, J. Endogenous ligands of TLR4 promote unresolving tissue fibrosis: Implications for systemic sclerosis and its targeted therapy. Immunol. Lett. 2018, 195, 9–17. [Google Scholar] [CrossRef] [PubMed]
  51. Moezinia, C.; Wong, V.; Watson, J.; Nagib, L.; Lopez Garces, S.; Zhang, S.; Ahmed Abdi, B.; Newton, F.; Abraham, D.; Stratton, R. Autoantibodies Which Bind to and Activate Keratinocytes in Systemic Sclerosis. Cells 2023, 12, 2490. [Google Scholar] [CrossRef]
  52. Zeng, C.; Kahlenberg, J.M.; Gudjonsson, J.E. IL-17A Softens the Skin: Antifibrotic Properties of IL-17A in Systemic Sclerosis. J. Investig. Dermatol. 2020, 140, 13–14. [Google Scholar] [CrossRef]
  53. Campochiaro, C.; Allanore, Y. An update on targeted therapies in systemic sclerosis based on a systematic review from the last 3 years. Arthritis Res. Ther. 2021, 23, 155. [Google Scholar] [CrossRef]
  54. Roofeh, D.; Lescoat, A.; Khanna, D. Emerging drugs for the treatment of scleroderma: A review of recent phase 2 and 3 trials. Expert. Opin. Emerg. Drugs 2020, 25, 455–466. [Google Scholar] [CrossRef]
  55. Connolly, M.K. Systemic sclerosis (scleroderma): Remaining challenges. Ann. Transl. Med. 2021, 9, 438. [Google Scholar] [CrossRef]
  56. Bukiri, H.; Volkmann, E.R. Current advances in the treatment of systemic sclerosis. Curr. Opin. Pharmacol. 2022, 64, 102211. [Google Scholar] [CrossRef]
  57. Ebata, S.; Yoshizaki-Ogawa, A.; Sato, S.; Yoshizaki, A. New Era in Systemic Sclerosis Treatment: Recently Approved Therapeutics. J. Clin. Med. 2022, 11, 4631. [Google Scholar] [CrossRef]
  58. Papadimitriou, T.I.; van Caam, A.; van der Kraan, P.M.; Thurlings, R.M. Therapeutic Options for Systemic Sclerosis: Current and Future Perspectives in Tackling Immune-Mediated Fibrosis. Biomedicines 2022, 10, 316. [Google Scholar] [CrossRef] [PubMed]
  59. Xue, E.; Minniti, A.; Alexander, T.; Del Papa, N.; Greco, R.; On Behalf of the Autoimmune Diseases Working Party (ADWP) of The European Society for Blood and Marrow Transplantation (EMBT). Cellular-Based Therapies in Systemic Sclerosis: From Hematopoietic Stem Cell Transplant to Innovative Approaches. Cells 2022, 11, 3346. [Google Scholar] [CrossRef]
  60. Farrell, J.; Ho, L. Management of Patients with Systemic Sclerosis-Associated Interstitial Lung Disease: A Focus on the Role of the Pharmacist. Integr. Pharm. Res. Pract. 2023, 12, 101–112. [Google Scholar] [CrossRef]
  61. Assassi, S.; Tumuluri, S.; Levin, R.W. Interstitial lung disease in patients with systemic sclerosis: What can we learn from the SENSCIS trial? Clin. Exp. Rheumatol. 2023, 41, 1713–1719. [Google Scholar] [CrossRef] [PubMed]
  62. Lescoat, A.; Roofeh, D.; Kuwana, M.; Lafyatis, R.; Allanore, Y.; Khanna, D. Therapeutic Approaches to Systemic Sclerosis: Recent Approvals and Future Candidate Therapies. Clin. Rev. Allergy Immunol. 2023, 64, 239–261. [Google Scholar] [CrossRef]
  63. Mulcaire-Jones, E.; Low, A.H.L.; Domsic, R.; Whitfield, M.L.; Khanna, D. Advances in biological and targeted therapies for systemic sclerosis. Expert. Opin. Biol. Ther. 2023, 23, 325–339. [Google Scholar] [CrossRef] [PubMed]
  64. Sen, C.K.; Gordillo, G.M.; Roy, S.; Kirsner, R.; Lambert, L.; Hunt, T.K.; Gottrup, F.; Gurtner, G.C.; Longaker, M.T. Human skin wounds: A major and snowballing threat to public health and the economy. Wound Repair Regen. 2009, 17, 763–771. [Google Scholar] [CrossRef]
  65. Sen, C.K. Human Wounds and Its Burden: An Updated Compendium of Estimates. Adv. Wound Care 2019, 8, 39–48. [Google Scholar] [CrossRef]
  66. Carter, M.J.; DaVanzo, J.; Haught, R.; Nusgart, M.; Cartwright, D.; Fife, C.E. Chronic wound prevalence and the associated cost of treatment in Medicare beneficiaries: Changes between 2014 and 2019. J. Med. Econ. 2023, 26, 894–901. [Google Scholar] [CrossRef] [PubMed]
  67. Nussbaum, S.R.; Carter, M.J.; Fife, C.E.; DaVanzo, J.; Haught, R.; Nusgart, M.; Cartwright, D. An Economic Evaluation of the Impact, Cost, and Medicare Policy Implications of Chronic Nonhealing Wounds. Value Health 2018, 21, 27–32. [Google Scholar] [CrossRef] [PubMed]
  68. Yang, G.; Cintina, I.; Pariser, A.; Oehrlein, E.; Sullivan, J.; Kennedy, A. The national economic burden of rare disease in the United States in 2019. Orphanet J. Rare Dis. 2022, 17, 163. [Google Scholar] [CrossRef]
  69. García-Pérez, L.; Linertová, R.; Valcárcel-Nazco, C.; Posada, M.; Gorostiza, I.; Serrano-Aguilar, P. Cost-of-illness studies in rare diseases: A scoping review. Orphanet J. Rare Dis. 2021, 16, 178. [Google Scholar] [CrossRef]
  70. Coffey, C.M.; Sandhu, A.S.; Crowson, C.S.; Achenbach, S.J.; Matteson, E.L.; Osborn, T.G.; Warrington, K.J.; Makol, A. Hospitalization Rates Are Highest in the First 5 Years of Systemic Sclerosis: Results from a Population-based Cohort (1980–2016). J. Rheumatol. 2021, 48, 877–882. [Google Scholar] [CrossRef]
  71. Fan, Y.; Bender, S.; Shi, W.; Zoz, D. Incidence and prevalence of systemic sclerosis and systemic sclerosis with interstitial lung disease in the United States. J. Manag. Care Spec. Pharm. 2020, 26, 1539–1547. [Google Scholar] [CrossRef]
  72. Khanna, D.; Furst, D.E.; Li, J.W.; Meng, Q.; Yuan, Y.; Lesperance, T.; Peoples, K.; Ali, F.; LaMoreaux, B.; Taylor, S.D. Economic and Health Care Resource Use Burden of Systemic Sclerosis. ACR Open Rheumatol. 2023, 5, 677–684. [Google Scholar] [CrossRef]
  73. Morrisroe, K.; Sandorfi, N.; Barron, M. Health Care Utilization: What Is the Cost of Caring for Scleroderma? Rheum. Dis. Clin. N. Am. 2023, 49, 359–375. [Google Scholar] [CrossRef]
  74. Colic, J.; Campochiaro, C.; Hughes, M.; Matucci Cerinic, M.; Dagna, L. Investigational drugs for the treatment of scleroderma: What’s new? Expert. Opin. Investig. Drugs 2023, 32, 601–614. [Google Scholar] [CrossRef]
  75. Farina, N.; Campochiaro, C.; Lescoat, A.; Benanti, G.; De Luca, G.; Khanna, D.; Dagna, L.; Matucci-Cerinic, M. Drug development and novel therapeutics to ensure a personalized approach in the treatment of systemic sclerosis. Expert. Rev. Clin. Immunol. 2023, 19, 1131–1142. [Google Scholar] [CrossRef]
  76. Komura, K.; Yanaba, K.; Bouaziz, J.D.; Yoshizaki, A.; Hasegawa, M.; Varga, J.; Takehara, K.; Matsushita, T. Perspective to precision medicine in scleroderma. Front. Immunol. 2023, 14, 1298665. [Google Scholar] [CrossRef] [PubMed]
  77. Lescoat, A.; Kato, H.; Varga, J. Emerging cellular and immunotherapies for systemic sclerosis: From mesenchymal stromal cells to CAR-T cells and vaccine-based approaches. Curr. Opin. Rheumatol. 2023, 35, 356–363. [Google Scholar] [CrossRef] [PubMed]
  78. Mascharak, S.; desJardins-Park, H.E.; Davitt, M.F.; Guardino, N.J.; Gurtner, G.C.; Wan, D.C.; Longaker, M.T. Modulating Cellular Responses to Mechanical Forces to Promote Wound Regeneration. Adv. Wound Care 2022, 11, 479–495. [Google Scholar] [CrossRef] [PubMed]
  79. Mascharak, S.; desJardins-Park, H.E.; Davitt, M.F.; Griffin, M.; Borrelli, M.R.; Moore, A.L.; Chen, K.; Duoto, B.; Chinta, M.; Foster, D.S.; et al. Preventing Engrailed-1 activation in fibroblasts yields wound regeneration without scarring. Science 2021, 372, eaba2374. [Google Scholar] [CrossRef]
  80. Yi, Z.; Zeng, J.; Chen, Z.; Chen, L.; Lu, H.B.; Zhang, Q.; Yang, X.; Qi, Z. The Role of Verteporfin in Prevention of Periprosthetic Capsular Fibrosis: An Experimental Study. Aesthet. Surg. J. 2022, 42, 820–829. [Google Scholar] [CrossRef]
  81. Wang, P.; Peng, Z.; Yu, L.; Liu, Y.; Wang, H.; Zhou, Z.; Liu, H.; Hong, S.; Nie, Y.; Deng, Y.; et al. Verteporfin-Loaded Bioadhesive Nanoparticles for the Prevention of Hypertrophic Scar. Small Methods 2024, 8, e2301295. [Google Scholar] [CrossRef]
  82. Berry, C.E.; Brenac, C.; Gonzalez, C.E.; Kendig, C.B.; Le, T.; An, N.; Griffin, M.F. Natural Compounds and Biomimetic Engineering to Influence Fibroblast Behavior in Wound Healing. Int. J. Mol. Sci. 2024, 25, 3274. [Google Scholar] [CrossRef]
  83. Cao, X.; Wu, X.; Zhang, Y.; Qian, X.; Sun, W.; Zhao, Y. Emerging biomedical technologies for scarless wound healing. Bioact. Mater. 2024, 42, 449–477. [Google Scholar] [CrossRef]
  84. Aebisher, D.; Szpara, J.; Bartusik-Aebisher, D. Advances in Medicine: Photodynamic Therapy. Int. J. Mol. Sci. 2024, 25, 8258. [Google Scholar] [CrossRef]
  85. Nong, J.; Shen, S.; Hong, F.; Xiao, F.; Meng, L.; Li, P.; Lei, X.; Chen, Y.G. Verteporfin inhibits TGF-β signaling by disrupting the Smad2/3-Smad4 interaction. Mol. Biol. Cell 2024, 35, ar95. [Google Scholar] [CrossRef]
  86. Chen, K.; Kwon, S.H.; Henn, D.; Kuehlmann, B.A.; Tevlin, R.; Bonham, C.A.; Griffin, M.; Trotsyuk, A.A.; Borrelli, M.R.; Noishiki, C.; et al. Disrupting biological sensors of force promotes tissue regeneration in large organisms. Nat. Commun. 2021, 12, 5256. [Google Scholar] [CrossRef]
  87. Bellamri, N.; Lelong, M.; Joannes, A.; Le Tallec, E.; Jouneau, S.; Vernhet, L.; Lescoat, A.; Lecureur, V. Effects of Ruxolitinib on fibrosis in preclinical models of systemic sclerosis. Int. Immunopharmacol. 2023, 116, 109723. [Google Scholar] [CrossRef]
  88. Moriana, C.; Moulinet, T.; Jaussaud, R.; Decker, P. JAK inhibitors and systemic sclerosis: A systematic review of the literature. Autoimmun. Rev. 2022, 21, 103168. [Google Scholar] [CrossRef]
  89. Spiera, R.; Kuwana, M.; Khanna, D.; Hummers, L.; Frech, T.M.; Stevens, W.; Matucci-Cerinic, M.; Kafaja, S.; Distler, O.; Jun, J.B.; et al. Efficacy and Safety of Lenabasum, a Cannabinoid Type 2 Receptor Agonist, in a Phase 3 Randomized Trial in Diffuse Cutaneous Systemic Sclerosis. Arthritis Rheumatol. 2023, 75, 1608–1618. [Google Scholar] [CrossRef] [PubMed]
  90. Burstein, S. Molecular Mechanisms for the Inflammation-Resolving Actions of Lenabasum. Mol. Pharmacol. 2021, 99, 125–132. [Google Scholar] [CrossRef]
  91. Werth, V.P.; Hejazi, E.; Pena, S.M.; Haber, J.; Zeidi, M.; Reddy, N.; Okawa, J.; Feng, R.; Bashir, M.M.; Gebre, K.; et al. Safety and Efficacy of Lenabasum, a Cannabinoid Receptor Type 2 Agonist, in Patients with Dermatomyositis with Refractory Skin Disease: A Randomized Clinical Trial. J. Investig. Dermatol. 2022, 142, 2651–2659.e2651. [Google Scholar] [CrossRef]
  92. Spiera, R.; Hummers, L.; Chung, L.; Frech, T.M.; Domsic, R.; Hsu, V.; Furst, D.E.; Gordon, J.; Mayes, M.; Simms, R.; et al. Safety and Efficacy of Lenabasum in a Phase II, Randomized, Placebo-Controlled Trial in Adults with Systemic Sclerosis. Arthritis Rheumatol. 2020, 72, 1350–1360. [Google Scholar] [CrossRef]
  93. Tian, N.; Cheng, H.; Du, Y.; Wang, X.; Lei, Y.; Liu, X.; Chen, M.; Xu, Z.; Wang, L.; Yin, H.; et al. Cannabinoid receptor 2 selective agonist alleviates systemic sclerosis by inhibiting Th2 differentiation through JAK/SOCS3 signaling. J. Autoimmun. 2024, 147, 103233. [Google Scholar] [CrossRef]
  94. Chen, Y.; Wu, L.; JHernández-Muñoz, J.; JMiller, M.; Pope, M.; Huyan, Y.; Zhong, L. The economic burden of systemic sclerosis—A systematic review. Int. J. Rheum. Dis. 2022, 25, 110–120. [Google Scholar] [CrossRef] [PubMed]
  95. Fullard, N.; Moles, A.; O’Reilly, S.; van Laar, J.M.; Faini, D.; Diboll, J.; Reynolds, N.J.; Mann, D.A.; Reichelt, J.; Oakley, F. The c-Rel subunit of NF-κB regulates epidermal homeostasis and promotes skin fibrosis in mice. Am. J. Pathol. 2013, 182, 2109–2120. [Google Scholar] [CrossRef]
  96. Giridharan, S.; Srinivasan, M. Mechanisms of NF-κB p65 and strategies for therapeutic manipulation. J. Inflamm. Res. 2018, 11, 407–419. [Google Scholar] [CrossRef] [PubMed]
  97. Micus, L.C.; Trautschold-Krause, F.S.; Jelit, A.L.; Schön, M.P.; Lorenz, V.N. NF-κB c-Rel modulates pre-fibrotic changes in human fibroblasts. Arch. Dermatol. Res. 2022, 314, 943–951. [Google Scholar] [CrossRef]
  98. Israël, A. The IKK complex, a central regulator of NF-kappaB activation. Cold Spring Harb. Perspect. Biol. 2010, 2, a000158. [Google Scholar] [CrossRef] [PubMed]
  99. Hinz, M.; Scheidereit, C. The IκB kinase complex in NF-κB regulation and beyond. EMBO Rep. 2014, 15, 46–61. [Google Scholar] [CrossRef]
  100. Heissmeyer, V.; Krappmann, D.; Hatada, E.N.; Scheidereit, C. Shared pathways of IkappaB kinase-induced SCF(betaTrCP)-mediated ubiquitination and degradation for the NF-kappaB precursor p105 and IkappaBalpha. Mol. Cell Biol. 2001, 21, 1024–1035. [Google Scholar] [CrossRef]
  101. Liu, T.; Zhang, L.; Joo, D.; Sun, S.C. NF-κB signaling in inflammation. Signal Transduct. Target. Ther. 2017, 2, 17023. [Google Scholar] [CrossRef]
  102. Gilmore, T.; Gapuzan, M.E.; Kalaitzidis, D.; Starczynowski, D. Rel/NF-kappa B/I kappa B signal transduction in the generation and treatment of human cancer. Cancer Lett. 2002, 181, 1–9. [Google Scholar] [CrossRef]
  103. Sun, S.C. Non-canonical NF-κB signaling pathway. Cell Res. 2011, 21, 71–85. [Google Scholar] [CrossRef]
  104. Moorthy, A.K.; Savinova, O.V.; Ho, J.Q.; Wang, V.Y.; Vu, D.; Ghosh, G. The 20S proteasome processes NF-kappaB1 p105 into p50 in a translation-independent manner. EMBO J. 2006, 25, 1945–1956. [Google Scholar] [CrossRef]
  105. Brignall, R.; Moody, A.T.; Mathew, S.; Gaudet, S. Considering Abundance, Affinity, and Binding Site Availability in the NF-κB Target Selection Puzzle. Front. Immunol. 2019, 10, 609. [Google Scholar] [CrossRef] [PubMed]
  106. Kumase, F.; Takeuchi, K.; Morizane, Y.; Suzuki, J.; Matsumoto, H.; Kataoka, K.; Al-Moujahed, A.; Maidana, D.E.; Miller, J.W.; Vavvas, D.G. AMPK-Activated Protein Kinase Suppresses Ccr2 Expression by Inhibiting the NF-κB Pathway in RAW264.7 Macrophages. PLoS ONE 2016, 11, e0147279. [Google Scholar] [CrossRef] [PubMed]
  107. Szołtysek, K.; Janus, P.; Zając, G.; Stokowy, T.; Walaszczyk, A.; Widłak, W.; Wojtaś, B.; Gielniewski, B.; Cockell, S.; Perkins, N.D.; et al. RRAD, IL4I1, CDKN1A, and SERPINE1 genes are potentially co-regulated by NF-κB and p53 transcription factors in cells exposed to high doses of ionizing radiation. BMC Genom. 2018, 19, 813. [Google Scholar] [CrossRef] [PubMed]
  108. Van Ginderachter, J.A. The wound healing chronicles. Blood 2012, 120, 499–500. [Google Scholar] [CrossRef] [PubMed]
  109. Schaffrick, L.; Ding, J.; Kwan, P.; Tredget, E. The dynamic changes of monocytes and cytokines during wound healing post-burn injury. Cytokine 2023, 168, 156231. [Google Scholar] [CrossRef]
  110. Boniakowski, A.E.; Kimball, A.S.; Joshi, A.; Schaller, M.; Davis, F.M.; denDekker, A.; Obi, A.T.; Moore, B.B.; Kunkel, S.L.; Gallagher, K.A. Murine macrophage chemokine receptor CCR2 plays a crucial role in macrophage recruitment and regulated inflammation in wound healing. Eur. J. Immunol. 2018, 48, 1445–1455. [Google Scholar] [CrossRef]
  111. Ågren, M.S.; Litman, T.; Eriksen, J.O.; Schjerling, P.; Bzorek, M.; Gjerdrum, L.M.R. Gene Expression Linked to Reepithelialization of Human Skin Wounds. Int. J. Mol. Sci. 2022, 23, 5746. [Google Scholar] [CrossRef]
  112. Kida, M.; Fatima, I.; Rozhkova, E.; Otero-Viñas, M.; Wu, M.; Kalin, J.H.; Cole, P.A.; Falanga, V.; Alani, R.M.; Sharov, A.A. Inhibition of the CoREST Repressor Complex Promotes Wound Re-Epithelialization through the Regulation of Keratinocyte Migration. J. Investig. Dermatol. 2024, 144, 378–386.e372. [Google Scholar] [CrossRef]
  113. Simone, T.M.; Higgins, C.E.; Czekay, R.P.; Law, B.K.; Higgins, S.P.; Archambeault, J.; Kutz, S.M.; Higgins, P.J. SERPINE1: A Molecular Switch in the Proliferation-Migration Dichotomy in Wound-”Activated” Keratinocytes. Adv. Wound Care 2014, 3, 281–290. [Google Scholar] [CrossRef]
  114. Ritsu, M.; Kawakami, K.; Kanno, E.; Tanno, H.; Ishii, K.; Imai, Y.; Maruyama, R.; Tachi, M. Critical role of tumor necrosis factor-α in the early process of wound healing in skin. J. Dermatol. Dermatol. Surg. 2017, 21, 14–19. [Google Scholar] [CrossRef]
  115. Frank, J.; Born, K.; Barker, J.H.; Marzi, I. In Vivo Effect of Tumor Necrosis Factor Alpha on Wound Angiogenesis and Epithelialization. Eur. J. Trauma. 2003, 29, 208–219. [Google Scholar] [CrossRef]
  116. Cao, Y.; Harvey, B.P.; Jin, L.; Westmoreland, S.; Wang, J.; Puri, M.; Yang, Y.; Robb, H.M.; Tanriverdi, S.; Hu, C.; et al. Therapeutic TNF Inhibitors Exhibit Differential Levels of Efficacy in Accelerating Cutaneous Wound Healing. JID Innov. 2024, 4, 100250. [Google Scholar] [CrossRef] [PubMed]
  117. Mihaly, S.R.; Ninomiya-Tsuji, J.; Morioka, S. TAK1 control of cell death. Cell Death Differ. 2014, 21, 1667–1676. [Google Scholar] [CrossRef] [PubMed]
  118. Martens, A.; van Loo, G. A20 at the Crossroads of Cell Death, Inflammation, and Autoimmunity. Cold Spring Harb. Perspect. Biol. 2020, 12, a036418. [Google Scholar] [CrossRef] [PubMed]
  119. Shamilov, R.; Aneskievich, B.J. TNIP1 in Autoimmune Diseases: Regulation of Toll-like Receptor Signaling. J. Immunol. Res. 2018, 2018, 3491269. [Google Scholar] [CrossRef]
  120. Zhou, J.; Rasmussen, N.L.; Olsvik, H.L.; Akimov, V.; Hu, Z.; Evjen, G.; Kaeser-Pebernard, S.; Sankar, D.S.; Roubaty, C.; Verlhac, P.; et al. TBK1 phosphorylation activates LIR-dependent degradation of the inflammation repressor TNIP1. J. Cell Biol. 2023, 222, e202108144. [Google Scholar] [CrossRef] [PubMed]
  121. Yamaguchi, K.; Shirakabe, K.; Shibuya, H.; Irie, K.; Oishi, I.; Ueno, N.; Taniguchi, T.; Nishida, E.; Matsumoto, K. Identification of a member of the MAPKKK family as a potential mediator of TGF-beta signal transduction. Science 1995, 270, 2008–2011. [Google Scholar] [CrossRef]
  122. Lee, J.; Mira-Arbibe, L.; Ulevitch, R.J. TAK1 regulates multiple protein kinase cascades activated by bacterial lipopolysaccharide. J. Leukoc. Biol. 2000, 68, 909–915. [Google Scholar] [CrossRef]
  123. Wang, W.; Gao, W.; Zhu, Q.; Alasbahi, A.; Seki, E.; Yang, L. TAK1: A Molecular Link Between Liver Inflammation, Fibrosis, Steatosis, and Carcinogenesis. Front. Cell Dev. Biol. 2021, 9, 734749. [Google Scholar] [CrossRef] [PubMed]
  124. Xu, Y.R.; Lei, C.Q. TAK1-TABs Complex: A Central Signalosome in Inflammatory Responses. Front. Immunol. 2020, 11, 608976. [Google Scholar] [CrossRef]
  125. Leask, A.; Naik, A.; Stratton, R.J. Back to the future: Targeting the extracellular matrix to treat systemic sclerosis. Nat. Rev. Rheumatol. 2023, 19, 713–723. [Google Scholar] [CrossRef]
  126. Landström, M. The TAK1-TRAF6 signalling pathway. Int. J. Biochem. Cell Biol. 2010, 42, 585–589. [Google Scholar] [CrossRef] [PubMed]
  127. Zhang, J.; Cao, L.; Lyu, L.; Qi, W.; Yang, W.; Ren, R.; Kao, C.; Zhang, Y.; Zhang, C.; Zhang, M. Regulation of TAK-TAB Complex Activation through Ubiquitylation. Front. Biosci. 2024, 29, 169. [Google Scholar] [CrossRef] [PubMed]
  128. Kanayama, A.; Seth, R.B.; Sun, L.; Ea, C.K.; Hong, M.; Shaito, A.; Chiu, Y.H.; Deng, L.; Chen, Z.J. TAB2 and TAB3 activate the NF-kappaB pathway through binding to polyubiquitin chains. Mol. Cell 2004, 15, 535–548. [Google Scholar] [CrossRef] [PubMed]
  129. Ge, B.; Gram, H.; Di Padova, F.; Huang, B.; New, L.; Ulevitch, R.J.; Luo, Y.; Han, J. MAPKK-independent activation of p38alpha mediated by TAB1-dependent autophosphorylation of p38alpha. Science 2002, 295, 1291–1294. [Google Scholar] [CrossRef]
  130. Sato, S.; Sanjo, H.; Takeda, K.; Ninomiya-Tsuji, J.; Yamamoto, M.; Kawai, T.; Matsumoto, K.; Takeuchi, O.; Akira, S. Essential function for the kinase TAK1 in innate and adaptive immune responses. Nat. Immunol. 2005, 6, 1087–1095. [Google Scholar] [CrossRef] [PubMed]
  131. Li, L.; Chen, Y.; Li, J.; Yin, H.; Guo, X.; Doan, J.; Molkentin, J.D.; Liu, Q. TAK1 Regulates Myocardial Response to Pathological Stress via NFAT, NFκB, and Bnip3 Pathways. Sci. Rep. 2015, 5, 16626. [Google Scholar] [CrossRef] [PubMed]
  132. Guo, F.; Hutchenreuther, J.; Carter, D.E.; Leask, A. TAK1 is required for dermal wound healing and homeostasis. J. Investig. Dermatol. 2013, 133, 1646–1654. [Google Scholar] [CrossRef]
  133. Maddali, P.; Ambesi, A.; McKeown-Longo, P.J. Induction of pro-inflammatory genes by fibronectin DAMPs in three fibroblast cell lines: Role of TAK1 and MAP kinases. PLoS ONE 2023, 18, e0286390. [Google Scholar] [CrossRef]
  134. Bale, S.; Verma, P.; Yalavarthi, B.; Scarneo, S.A.; Hughes, P.; Amin, M.A.; Tsou, P.S.; Khanna, D.; Haystead, T.A.; Bhattacharyya, S.; et al. Pharmacological inhibition of TAK1 prevents and induces regression of experimental organ fibrosis. JCI Insight 2023, 8, e165358. [Google Scholar] [CrossRef]
  135. Freeze, R.; Yang, K.W.; Haystead, T.; Hughes, P.; Scarneo, S. Delineation of the distinct inflammatory signaling roles of TAK1 and JAK1/3 in the CIA model of rheumatoid arthritis. Pharmacol. Res. Perspect. 2023, 11, e01124. [Google Scholar] [CrossRef]
  136. Asano, Y.; Varga, J. Rationally-based therapeutic disease modification in systemic sclerosis: Novel strategies. Semin. Cell Dev. Biol. 2020, 101, 146–160. [Google Scholar] [CrossRef] [PubMed]
  137. Totzke, J.; Scarneo, S.A.; Yang, K.W.; Haystead, T.A.J. TAK1: A potent tumour necrosis factor inhibitor for the treatment of inflammatory diseases. Open Biol. 2020, 10, 200099. [Google Scholar] [CrossRef] [PubMed]
  138. Scarneo, S.; Hughes, P.; Freeze, R.; Yang, K.; Totzke, J.; Haystead, T. Development and Efficacy of an Orally Bioavailable Selective TAK1 Inhibitor for the Treatment of Inflammatory Arthritis. ACS Chem. Biol. 2022, 17, 536–544. [Google Scholar] [CrossRef] [PubMed]
  139. Lamontagne, N. EydisBio’s Systemic Sclerosis Treatment Receives FDA Orphan Drug Designation. Available online: https://www.ncbiotech.org/news/eydisbios-systemic-sclerosis-treatment-receives-fda-orphan-drug-designation (accessed on 20 November 2024).
  140. Cui, H.S.; Joo, S.Y.; Cho, Y.S.; Kim, J.B.; Seo, C.H. CPEB1 or CPEB4 knockdown suppresses the TAK1 and Smad signalings in THP-1 macrophage-like cells and dermal fibroblasts. Arch. Biochem. Biophys. 2020, 683, 108322. [Google Scholar] [CrossRef] [PubMed]
  141. Woeller, C.F.; O’Loughlin, C.W.; Roztocil, E.; Feldon, S.E.; Phipps, R.P. Salinomycin and other polyether ionophores are a new class of antiscarring agent. J. Biol. Chem. 2015, 290, 3563–3575. [Google Scholar] [CrossRef]
  142. Fang, Q.Q.; Wang, X.F.; Zhao, W.Y.; Ding, S.L.; Shi, B.H.; Xia, Y.; Yang, H.; Wu, L.H.; Li, C.Y.; Tan, W.Q. Angiotensin-converting enzyme inhibitor reduces scar formation by inhibiting both canonical and noncanonical TGF-β1 pathways. Sci. Rep. 2018, 8, 3332. [Google Scholar] [CrossRef]
  143. Cui, H.S.; Lee, Y.R.; Ro, Y.M.; Joo, S.Y.; Cho, Y.S.; Kim, J.B.; Kim, D.H.; Seo, C.H. Knockdown of CPEB1 and CPEB4 Inhibits Scar Formation via Modulation of TAK1 and SMAD Signaling. Ann. Dermatol. 2023, 35, 293–302. [Google Scholar] [CrossRef] [PubMed]
  144. Cui, H.S.; Kim, D.H.; Joo, S.Y.; Cho, Y.S.; Kim, J.B.; Seo, C.H. Exosomes derived from human hypertrophic scar fibroblasts induces smad and TAK1 signaling in normal dermal fibroblasts. Arch. Biochem. Biophys. 2022, 722, 109215. [Google Scholar] [CrossRef]
  145. Dixit, V.M.; Green, S.; Sarma, V.; Holzman, L.B.; Wolf, F.W.; O’Rourke, K.; Ward, P.A.; Prochownik, E.V.; Marks, R.M. Tumor necrosis factor-alpha induction of novel gene products in human endothelial cells including a macrophage-specific chemotaxin. J. Biol. Chem. 1990, 265, 2973–2978. [Google Scholar] [CrossRef]
  146. Catrysse, L.; Vereecke, L.; Beyaert, R.; van Loo, G. A20 in inflammation and autoimmunity. Trends Immunol. 2014, 35, 22–31. [Google Scholar] [CrossRef]
  147. Wertz, I.E.; O’Rourke, K.M.; Zhou, H.; Eby, M.; Aravind, L.; Seshagiri, S.; Wu, P.; Wiesmann, C.; Baker, R.; Boone, D.L.; et al. De-ubiquitination and ubiquitin ligase domains of A20 downregulate NF-kappaB signalling. Nature 2004, 430, 694–699. [Google Scholar] [CrossRef] [PubMed]
  148. Sun, S.C. A20 restricts inflammation via ubiquitin binding. Nat. Immunol. 2020, 21, 362–364. [Google Scholar] [CrossRef] [PubMed]
  149. Heyninck, K.; De Valck, D.; Vanden Berghe, W.; Van Criekinge, W.; Contreras, R.; Fiers, W.; Haegeman, G.; Beyaert, R. The zinc finger protein A20 inhibits TNF-induced NF-kappaB-dependent gene expression by interfering with an RIP- or TRAF2-mediated transactivation signal and directly binds to a novel NF-kappaB-inhibiting protein ABIN. J. Cell Biol. 1999, 145, 1471–1482. [Google Scholar] [CrossRef] [PubMed]
  150. Polykratis, A.; Martens, A.; Eren, R.O.; Shirasaki, Y.; Yamagishi, M.; Yamaguchi, Y.; Uemura, S.; Miura, M.; Holzmann, B.; Kollias, G.; et al. A20 prevents inflammasome-dependent arthritis by inhibiting macrophage necroptosis through its ZnF7 ubiquitin-binding domain. Nat. Cell Biol. 2019, 21, 731–742. [Google Scholar] [CrossRef] [PubMed]
  151. Nanda, S.K.; Venigalla, R.K.; Ordureau, A.; Patterson-Kane, J.C.; Powell, D.W.; Toth, R.; Arthur, J.S.; Cohen, P. Polyubiquitin binding to ABIN1 is required to prevent autoimmunity. J. Exp. Med. 2011, 208, 1215–1228. [Google Scholar] [CrossRef] [PubMed]
  152. Wagner, S.; Carpentier, I.; Rogov, V.; Kreike, M.; Ikeda, F.; Löhr, F.; Wu, C.J.; Ashwell, J.D.; Dötsch, V.; Dikic, I.; et al. Ubiquitin binding mediates the NF-kappaB inhibitory potential of ABIN proteins. Oncogene 2008, 27, 3739–3745. [Google Scholar] [CrossRef] [PubMed]
  153. Boudra, R.; Ramsey, M.R. Understanding Transcriptional Networks Regulating Initiation of Cutaneous Wound Healing. Yale J. Biol. Med. 2020, 93, 161–173. [Google Scholar]
  154. Dong, J.; Ma, Q. In Vivo Activation and Pro-Fibrotic Function of NF-κB in Fibroblastic Cells During Pulmonary Inflammation and Fibrosis Induced by Carbon Nanotubes. Front. Pharmacol. 2019, 10, 1140. [Google Scholar] [CrossRef]
  155. Wullaert, A.; Bonnet, M.C.; Pasparakis, M. NF-κB in the regulation of epithelial homeostasis and inflammation. Cell Res. 2011, 21, 146–158. [Google Scholar] [CrossRef]
  156. Wang, W.; Bale, S.; Wei, J.; Yalavarthi, B.; Bhattacharyya, D.; Yan, J.J.; Abdala-Valencia, H.; Xu, D.; Sun, H.; Marangoni, R.G.; et al. Fibroblast A20 governs fibrosis susceptibility and its repression by DREAM promotes fibrosis in multiple organs. Nat. Commun. 2022, 13, 6358. [Google Scholar] [CrossRef] [PubMed]
  157. Dieudé, P.; Guedj, M.; Wipff, J.; Ruiz, B.; Riemekasten, G.; Matucci-Cerinic, M.; Melchers, I.; Hachulla, E.; Airo, P.; Diot, E.; et al. Association of the TNFAIP3 rs5029939 variant with systemic sclerosis in the European Caucasian population. Ann. Rheum. Dis. 2010, 69, 1958–1964. [Google Scholar] [CrossRef] [PubMed]
  158. Martin, J.E.; Assassi, S.; Diaz-Gallo, L.M.; Broen, J.C.; Simeon, C.P.; Castellvi, I.; Vicente-Rabaneda, E.; Fonollosa, V.; Ortego-Centeno, N.; González-Gay, M.A.; et al. A systemic sclerosis and systemic lupus erythematosus pan-meta-GWAS reveals new shared susceptibility loci. Hum. Mol. Genet. 2013, 22, 4021–4029. [Google Scholar] [CrossRef] [PubMed]
  159. Martin, J.E.; Broen, J.C.; Carmona, F.D.; Teruel, M.; Simeon, C.P.; Vonk, M.C.; van ‘t Slot, R.; Rodriguez-Rodriguez, L.; Vicente, E.; Fonollosa, V.; et al. Identification of CSK as a systemic sclerosis genetic risk factor through Genome Wide Association Study follow-up. Hum. Mol. Genet. 2012, 21, 2825–2835. [Google Scholar] [CrossRef] [PubMed]
  160. Terao, C.; Ohmura, K.; Kawaguchi, Y.; Nishimoto, T.; Kawasaki, A.; Takehara, K.; Furukawa, H.; Kochi, Y.; Ota, Y.; Ikari, K.; et al. PLD4 as a novel susceptibility gene for systemic sclerosis in a Japanese population. Arthritis Rheum. 2013, 65, 472–480. [Google Scholar] [CrossRef] [PubMed]
  161. Gorlova, O.Y.; Li, Y.; Gorlov, I.; Ying, J.; Chen, W.V.; Assassi, S.; Reveille, J.D.; Arnett, F.C.; Zhou, X.; Bossini-Castillo, L.; et al. Gene-level association analysis of systemic sclerosis: A comparison of African-Americans and White populations. PLoS ONE 2018, 13, e0189498. [Google Scholar] [CrossRef]
  162. Ota, Y.; Kuwana, M. Updates on genetics in systemic sclerosis. Inflamm. Regen. 2021, 41, 17. [Google Scholar] [CrossRef]
  163. Wei, P.; Yang, Y.; Guo, X.; Hei, N.; Lai, S.; Assassi, S.; Liu, M.; Tan, F.; Zhou, X. Identification of an Association of TNFAIP3 Polymorphisms with Matrix Metalloproteinase Expression in Fibroblasts in an Integrative Study of Systemic Sclerosis-Associated Genetic and Environmental Factors. Arthritis Rheumatol. 2016, 68, 749–760. [Google Scholar] [CrossRef] [PubMed]
  164. Malcomson, B.; Wilson, H.; Veglia, E.; Thillaiyampalam, G.; Barsden, R.; Donegan, S.; El Banna, A.; Elborn, J.S.; Ennis, M.; Kelly, C.; et al. Connectivity mapping (ssCMap) to predict A20-inducing drugs and their antiinflammatory action in cystic fibrosis. Proc. Natl. Acad. Sci. USA 2016, 113, E3725–E3734. [Google Scholar] [CrossRef]
  165. Reihill, J.A.; Malcomson, B.; Bertelsen, A.; Cheung, S.; Czerwiec, A.; Barsden, R.; Elborn, J.S.; Dürkop, H.; Hirsch, B.; Ennis, M.; et al. Induction of the inflammatory regulator A20 by gibberellic acid in airway epithelial cells. Br. J. Pharmacol. 2016, 173, 778–789. [Google Scholar] [CrossRef]
  166. Han, M.T.; Pei, H.; Sun, Q.Q.; Wang, C.L.; Li, P.; Xie, Y.Y.; Cao, L.J.; Zhang, X.X.; Sun, Z.L. ZY-444 inhibits the growth and metastasis of prostate cancer by targeting TNFAIP3 through TNF signaling pathway. Am. J. Cancer Res. 2023, 13, 1533–1546. [Google Scholar]
  167. Fang, F.; Liu, L.; Yang, Y.; Tamaki, Z.; Wei, J.; Marangoni, R.G.; Bhattacharyya, S.; Summer, R.S.; Ye, B.; Varga, J. The adipokine adiponectin has potent anti-fibrotic effects mediated via adenosine monophosphate-activated protein kinase: Novel target for fibrosis therapy. Arthritis Res. Ther. 2012, 14, R229. [Google Scholar] [CrossRef] [PubMed]
  168. Wei, J.; Fang, F.; Lam, A.P.; Sargent, J.L.; Hamburg, E.; Hinchcliff, M.E.; Gottardi, C.J.; Atit, R.; Whitfield, M.L.; Varga, J. Wnt/β-catenin signaling is hyperactivated in systemic sclerosis and induces Smad-dependent fibrotic responses in mesenchymal cells. Arthritis Rheum. 2012, 64, 2734–2745. [Google Scholar] [CrossRef] [PubMed]
  169. Rusu, I.; Mennillo, E.; Bain, J.L.; Li, Z.; Sun, X.; Ly, K.M.; Rosli, Y.Y.; Naser, M.; Wang, Z.; Advincula, R.; et al. Microbial signals, MyD88, and lymphotoxin drive TNF-independent intestinal epithelial tissue damage. J. Clin. Investig. 2022, 132, e154993. [Google Scholar] [CrossRef] [PubMed]
  170. Harirchian, P.; Lee, J.; Hilz, S.; Sedgewick, A.J.; Perez White, B.E.; Kesling, M.J.; Mully, T.; Golovato, J.; Gray, M.; Mauro, T.M.; et al. A20 and ABIN1 Suppression of a Keratinocyte Inflammatory Program with a Shared Single-Cell Expression Signature in Diverse Human Rashes. J. Investig. Dermatol. 2019, 139, 1264–1273. [Google Scholar] [CrossRef] [PubMed]
  171. Kattah, M.G.; Shao, L.; Rosli, Y.Y.; Shimizu, H.; Whang, M.I.; Advincula, R.; Achacoso, P.; Shah, S.; Duong, B.H.; Onizawa, M.; et al. A20 and ABIN-1 synergistically preserve intestinal epithelial cell survival. J. Exp. Med. 2018, 215, 1839–1852. [Google Scholar] [CrossRef]
  172. Shamilov, R.; Ackley, T.W.; Aneskievich, B.J. Enhanced Wound Healing- and Inflammasome-Associated Gene Expression in TNFAIP3-Interacting Protein 1- (TNIP1-) Deficient HaCaT Keratinocytes Parallels Reduced Reepithelialization. Mediat. Inflamm. 2020, 2020, 5919150. [Google Scholar] [CrossRef] [PubMed]
  173. Dziedzic, S.A.; Su, Z.; Jean Barrett, V.; Najafov, A.; Mookhtiar, A.K.; Amin, P.; Pan, H.; Sun, L.; Zhu, H.; Ma, A.; et al. ABIN-1 regulates RIPK1 activation by linking Met1 ubiquitylation with Lys63 deubiquitylation in TNF-RSC. Nat. Cell Biol. 2018, 20, 58–68. [Google Scholar] [CrossRef]
  174. Kuriakose, J.; Redecke, V.; Guy, C.; Zhou, J.; Wu, R.; Ippagunta, S.K.; Tillman, H.; Walker, P.D.; Vogel, P.; Häcker, H. Patrolling monocytes promote the pathogenesis of early lupus-like glomerulonephritis. J. Clin. Investig. 2019, 129, 2251–2265. [Google Scholar] [CrossRef]
  175. G’Sell, R.T.; Gaffney, P.M.; Powell, D.W. A20-Binding Inhibitor of NF-κB Activation 1 is a Physiologic Inhibitor of NF-κB: A Molecular Switch for Inflammation and Autoimmunity. Arthritis Rheumatol. 2015, 67, 2292–2302. [Google Scholar] [CrossRef]
  176. Oshima, S.; Turer, E.E.; Callahan, J.A.; Chai, S.; Advincula, R.; Barrera, J.; Shifrin, N.; Lee, B.; Benedict Yen, T.S.; Woo, T.; et al. ABIN-1 is a ubiquitin sensor that restricts cell death and sustains embryonic development. Nature 2009, 457, 906–909. [Google Scholar] [CrossRef]
  177. Verstrepen, L.; Carpentier, I.; Verhelst, K.; Beyaert, R. ABINs: A20 binding inhibitors of NF-kappa B and apoptosis signaling. Biochem. Pharmacol. 2009, 78, 105–114. [Google Scholar] [CrossRef] [PubMed]
  178. Korte, E.A.; Caster, D.J.; Barati, M.T.; Tan, M.; Zheng, S.; Berthier, C.C.; Brosius, F.C., 3rd; Vieyra, M.B.; Sheehan, R.M.; Kosiewicz, M.; et al. ABIN1 Determines Severity of Glomerulonephritis via Activation of Intrinsic Glomerular Inflammation. Am. J. Pathol. 2017, 187, 2799–2810. [Google Scholar] [CrossRef] [PubMed]
  179. Li, M.; Liu, Y.; Xu, C.; Zhao, Q.; Liu, J.; Xing, M.; Li, X.; Zhang, H.; Wu, X.; Wang, L.; et al. Ubiquitin-binding domain in ABIN1 is critical for regulating cell death and inflammation during development. Cell Death Differ. 2022, 29, 2034–2045. [Google Scholar] [CrossRef] [PubMed]
  180. Allanore, Y.; Saad, M.; Dieudé, P.; Avouac, J.; Distler, J.H.; Amouyel, P.; Matucci-Cerinic, M.; Riemekasten, G.; Airo, P.; Melchers, I.; et al. Genome-wide scan identifies TNIP1, PSORS1C1, and RHOB as novel risk loci for systemic sclerosis. PLoS Genet. 2011, 7, e1002091. [Google Scholar] [CrossRef]
  181. Orvain, C.; Assassi, S.; Avouac, J.; Allanore, Y. Systemic sclerosis pathogenesis: Contribution of recent advances in genetics. Curr. Opin. Rheumatol. 2020, 32, 505–514. [Google Scholar] [CrossRef]
  182. López-Isac, E.; Acosta-Herrera, M.; Kerick, M.; Assassi, S.; Satpathy, A.T.; Granja, J.; Mumbach, M.R.; Beretta, L.; Simeón, C.P.; Carreira, P.; et al. GWAS for systemic sclerosis identifies multiple risk loci and highlights fibrotic and vasculopathy pathways. Nat. Commun. 2019, 10, 4955. [Google Scholar] [CrossRef]
  183. Bossini-Castillo, L.; Martin, J.E.; Broen, J.; Simeon, C.P.; Beretta, L.; Gorlova, O.Y.; Vonk, M.C.; Ortego-Centeno, N.; Espinosa, G.; Carreira, P.; et al. Confirmation of TNIP1 but not RHOB and PSORS1C1 as systemic sclerosis risk factors in a large independent replication study. Ann. Rheum. Dis. 2013, 72, 602–607. [Google Scholar] [CrossRef] [PubMed]
  184. Li, T.; Ortiz-Fernández, L.; Andrés-León, E.; Ciudad, L.; Javierre, B.M.; López-Isac, E.; Guillén-Del-Castillo, A.; Simeón-Aznar, C.P.; Ballestar, E.; Martin, J. Epigenomics and transcriptomics of systemic sclerosis CD4+ T cells reveal long-range dysregulation of key inflammatory pathways mediated by disease-associated susceptibility loci. Genome Med. 2020, 12, 81. [Google Scholar] [CrossRef]
  185. Ortíz-Fernández, L.; Martín, J.; Alarcón-Riquelme, M.E. A Summary on the Genetics of Systemic Lupus Erythematosus, Rheumatoid Arthritis, Systemic Sclerosis, and Sjögren’s Syndrome. Clin. Rev. Allergy Immunol. 2023, 64, 392–411. [Google Scholar] [CrossRef]
  186. Li, X.; Ampleford, E.J.; Howard, T.D.; Moore, W.C.; Torgerson, D.G.; Li, H.; Busse, W.W.; Castro, M.; Erzurum, S.C.; Israel, E.; et al. Genome-wide association studies of asthma indicate opposite immunopathogenesis direction from autoimmune diseases. J. Allergy Clin. Immunol. 2012, 130, 861–868.e867. [Google Scholar] [CrossRef]
  187. Milano, A.; Pendergrass, S.A.; Sargent, J.L.; George, L.K.; McCalmont, T.H.; Connolly, M.K.; Whitfield, M.L. Molecular subsets in the gene expression signatures of scleroderma skin. PLoS ONE 2008, 3, e2696. [Google Scholar] [CrossRef]
  188. Xiong, H.Y.; Alipanahi, B.; Lee, L.J.; Bretschneider, H.; Merico, D.; Yuen, R.K.; Hua, Y.; Gueroussov, S.; Najafabadi, H.S.; Hughes, T.R.; et al. RNA splicing. The human splicing code reveals new insights into the genetic determinants of disease. Science 2015, 347, 1254806. [Google Scholar] [CrossRef]
  189. Cruz, J.A.; Childs, E.E.; Amatya, N.; Garg, A.V.; Beyaert, R.; Kane, L.P.; Aneskievich, B.J.; Ma, A.; Gaffen, S.L. Interleukin-17 signaling triggers degradation of the constitutive NF-κB inhibitor ABIN-1. Immunohorizons 2017, 1, 133–141. [Google Scholar] [CrossRef] [PubMed]
  190. Shinkawa, Y.; Imami, K.; Fuseya, Y.; Sasaki, K.; Ohmura, K.; Ishihama, Y.; Morinobu, A.; Iwai, K. ABIN1 is a signal-induced autophagy receptor that attenuates NF-κB activation by recognizing linear ubiquitin chains. FEBS Lett. 2022, 596, 1147–1164. [Google Scholar] [CrossRef] [PubMed]
  191. Sharma, A.; Sharma, D.; Zhao, F. Updates on Recent Clinical Assessment of Commercial Chronic Wound Care Products. Adv. Healthc. Mater. 2023, 12, e2300556. [Google Scholar] [CrossRef] [PubMed]
  192. Deng, X.; Gould, M.; Ali, M.A. A review of current advancements for wound healing: Biomaterial applications and medical devices. J. Biomed. Mater. Res. B Appl. Biomater. 2022, 110, 2542–2573. [Google Scholar] [CrossRef] [PubMed]
  193. Dai, X.; Shiraishi, K.; Muto, J.; Mori, H.; Murakami, M.; Sayama, K. Nuclear IL-33 Plays an Important Role in EGFR-Mediated Keratinocyte Migration by Regulating the Activation of Signal Transducer and Activator of Transcription 3 and NF-κB. JID Innov. 2023, 3, 100205. [Google Scholar] [CrossRef]
  194. Chang, M.; Nguyen, T.T. Strategy for Treatment of Infected Diabetic Foot Ulcers. Acc. Chem. Res. 2021, 54, 1080–1093. [Google Scholar] [CrossRef]
  195. Wang, B.; Zhang, J.; Li, G.; Xu, C.; Yang, L.; Wu, Y.; Liu, Y.; Liu, Z.; Wang, M.; Li, J.; et al. N-acetyltransferase 10 promotes cutaneous wound repair via the NF-κB-IL-6 axis. Cell Death Discov. 2023, 9, 324. [Google Scholar] [CrossRef]
  196. Zhu, H.; Li, Q.; Huang, Q.; Yang, H.; Zheng, J.; Xie, R.; Han, D.; Wei, Q. RIG-I contributes to keratinocyte proliferation and wound repair by inducing TIMP-1 expression through NF-κB signaling pathway. J. Cell Physiol. 2023, 238, 1876–1890. [Google Scholar] [CrossRef]
  197. Dolivo, D.M.; Sun, L.S.; Rodrigues, A.E.; Galiano, R.D.; Mustoe, T.A.; Hong, S.J. Epidermal Potentiation of Dermal Fibrosis: Lessons from Occlusion and Mucosal Healing. Am. J. Pathol. 2023, 193, 510–519. [Google Scholar] [CrossRef]
  198. Aden, N.; Shiwen, X.; Aden, D.; Black, C.; Nuttall, A.; Denton, C.P.; Leask, A.; Abraham, D.; Stratton, R. Proteomic analysis of scleroderma lesional skin reveals activated wound healing phenotype of epidermal cell layer. Rheumatology 2008, 47, 1754–1760. [Google Scholar] [CrossRef]
  199. Sarate, R.M.; Hochstetter, J.; Valet, M.; Hallou, A.; Song, Y.; Bansaccal, N.; Ligare, M.; Aragona, M.; Engelman, D.; Bauduin, A.; et al. Dynamic regulation of tissue fluidity controls skin repair during wound healing. Cell 2024, 187, 5298–5315.e5219. [Google Scholar] [CrossRef] [PubMed]
  200. Xie, J.; Wu, X.; Zheng, S.; Lin, K.; Su, J. Aligned electrospun poly(L-lactide) nanofibers facilitate wound healing by inhibiting macrophage M1 polarization via the JAK-STAT and NF-κB pathways. J. Nanobiotechnol. 2022, 20, 342. [Google Scholar] [CrossRef] [PubMed]
  201. Tu, Z.; Zhong, Y.; Hu, H.; Shao, D.; Haag, R.; Schirner, M.; Lee, J.; Sullenger, B.; Leong, K.W. Design of therapeutic biomaterials to control inflammation. Nat. Rev. Mater. 2022, 7, 557–574. [Google Scholar] [CrossRef] [PubMed]
  202. Shaabani, E.; Sharifiaghdam, M.; Faridi-Majidi, R.; De Smedt, S.C.; Braeckmans, K.; Fraire, J.C. Gene therapy to enhance angiogenesis in chronic wounds. Mol. Ther. Nucleic Acids 2022, 29, 871–899. [Google Scholar] [CrossRef] [PubMed]
  203. Yuan, T.; Tang, H.; Xu, X.; Shao, J.; Wu, G.; Cho, Y.C.; Ping, Y.; Liang, G. Inflammation conditional genome editing mediated by the CRISPR-Cas9 system. iScience 2023, 26, 106872. [Google Scholar] [CrossRef] [PubMed]
  204. Li, J.; Wei, M.; Liu, X.; Xiao, S.; Cai, Y.; Li, F.; Tian, J.; Qi, F.; Xu, G.; Deng, C. The progress, prospects, and challenges of the use of non-coding RNA for diabetic wounds. Mol. Ther. Nucleic Acids 2021, 24, 554–578. [Google Scholar] [CrossRef]
  205. Dikic, I.; Schulman, B.A. An expanded lexicon for the ubiquitin code. Nat. Rev. Mol. Cell Biol. 2023, 24, 273–287. [Google Scholar] [CrossRef]
  206. Akizuki, Y.; Kaypee, S.; Ohtake, F.; Ikeda, F. The emerging roles of non-canonical ubiquitination in proteostasis and beyond. J. Cell Biol. 2024, 223, e202311171. [Google Scholar] [CrossRef] [PubMed]
  207. Pan, H.; Song, J.; An, Q.; Chen, J.; Zheng, W.; Zhang, L.; Gu, J.; Deng, C.; Yang, B. Inhibition of Ubiquitin C-Terminal Hydrolase L1 Facilitates Cutaneous Wound Healing via Activating TGF-β/Smad Signalling Pathway in Fibroblasts. Exp. Dermatol. 2024, 33, e15186. [Google Scholar] [CrossRef] [PubMed]
Figure 1. TAK1, TNFAIP3, and TNIP1 are focal point for regulating activation of NF-κB, leading to wound healing and fibrotic disorder gene expression (see text). In brief, function of TAK1 leads to progression of cytoplasmic signaling downstream of the indicated receptors, while TNFAIP3 and TNIP1 restrict continuation of the signal. Created in Biorender.com.
Figure 1. TAK1, TNFAIP3, and TNIP1 are focal point for regulating activation of NF-κB, leading to wound healing and fibrotic disorder gene expression (see text). In brief, function of TAK1 leads to progression of cytoplasmic signaling downstream of the indicated receptors, while TNFAIP3 and TNIP1 restrict continuation of the signal. Created in Biorender.com.
Biomedicines 12 02723 g001
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

Samulevich, M.L.; Carman, L.E.; Aneskievich, B.J. Critical Analysis of Cytoplasmic Progression of Inflammatory Signaling Suggests Potential Pharmacologic Targets for Wound Healing and Fibrotic Disorders. Biomedicines 2024, 12, 2723. https://doi.org/10.3390/biomedicines12122723

AMA Style

Samulevich ML, Carman LE, Aneskievich BJ. Critical Analysis of Cytoplasmic Progression of Inflammatory Signaling Suggests Potential Pharmacologic Targets for Wound Healing and Fibrotic Disorders. Biomedicines. 2024; 12(12):2723. https://doi.org/10.3390/biomedicines12122723

Chicago/Turabian Style

Samulevich, Michael L., Liam E. Carman, and Brian J. Aneskievich. 2024. "Critical Analysis of Cytoplasmic Progression of Inflammatory Signaling Suggests Potential Pharmacologic Targets for Wound Healing and Fibrotic Disorders" Biomedicines 12, no. 12: 2723. https://doi.org/10.3390/biomedicines12122723

APA Style

Samulevich, M. L., Carman, L. E., & Aneskievich, B. J. (2024). Critical Analysis of Cytoplasmic Progression of Inflammatory Signaling Suggests Potential Pharmacologic Targets for Wound Healing and Fibrotic Disorders. Biomedicines, 12(12), 2723. https://doi.org/10.3390/biomedicines12122723

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