IMR Press / FBL / Volume 27 / Issue 1 / DOI: 10.31083/j.fbl2701031
Open Access Systematic Review
Isoform-specific and cell/tissue-dependent effects of p38 MAPKs in regulating inflammation and inflammation-associated oncogenesis
Show Less
1 Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53223, USA
2 Research Service, Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, WI 53295, USA
*Correspondence: gchen@mcw.edu (Guan Chen)
Academic Editor: Graham Pawelec
Front. Biosci. (Landmark Ed) 2022, 27(1), 31; https://doi.org/10.31083/j.fbl2701031
Submitted: 11 November 2021 | Revised: 29 December 2021 | Accepted: 29 December 2021 | Published: 18 January 2022
Copyright: © 2022 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

p38 MAPK (mitogen-activated protein kinases) family proteins (α, β, γ and δ) are key inflammatory kinases and play an important role in relaying and processing intrinsic and extrinsic signals in response to inflammation, stress, and oncogene to regulate cell growth, cell death and cell transformation. Recent studies in genetic mouse models revealed that p38α in epithelial cells mostly suppresses whereas in immune cells it promotes inflammation and inflammation-associated oncogenesis. On the contrary, p38γ and p38δ signaling in immune and epithelial cells is both pro-inflammatory and oncogenic. This review summarizes recent discoveries in this field, discusses possible associated mechanisms, and highlights potentials of systemically targeting isoform-specific p38 MAPKs. Understanding of p38 MAPK isoform-specific and cell/tissue- and perhaps stage-dependent effects and their integrated regulated activity in inflammation and in inflammation-associated oncogenesis is essential for effectively targeting this group of kinases for therapeutic intervention.

Keywords
p38 MAPKs
Isoform-specific and cell/tissue-dependent effects
Inflammation
Inflammation-associated oncogenesis
1. Introduction

p38 mitogen-activated protein kinases (MAPKs) (α, β, γ and δ) are encoded by four different genes in four different chromosomes [1]. p38 MAPKs are dual-phosphorylated on tyrosine and threonine residues within a conserved Thr-Pro-Tyr (TPY) motif by MAPK kinase 3 (MKK3) and/or MKK6, which in turn phosphorylate a substrate typically containing a ST/P motif (Ser or Thr residue, followed by Pro [1]). p38α and p38β phosphorylate more than 100 substrates [2], and many of them are not phosphorylated by p38γ and p38δ that have specific and non-overlapping substrates and are therefore called alternative p38 MAPKs [3, 4, 5]. Although distinct substrates may play a role in an isoform-specific effect of p38 MAPKs, how p38 MAPK family members signal via common and unique substrates are largely unknown [2, 4]. We will review recent discoveries from genetic studies about isoform-specific and cell/tissue-dependent effects of p38 MAPKs in inflammation and in inflammation-associated oncogenesis and discuss potentials of targeting a specific p38 isoform in therapeutic intervention.

p38α is expressed universally in all tissues and/or cells, whereas other p38 family proteins are only detectable in certain tissues and/or cells [1, 2]. Although all p38 MAPKs can be activated similarly in response to inflammation, stress and oncogenic signaling, they can also be activated distinctively [1, 2, 6, 7, 8, 9]. Oncogene RAS, for example, stimulates p38α (also called p38) phosphorylation but increases RNA/protein levels of p38γ (and not other p38 MAPKs), indicating that p38 MAPKs are activated by Ras oncogene by an isoform-specific mechanism [6, 7, 10, 11, 12]. Furthermore, elevated p38γ gene expression was demonstrated in human breast, colon, and pancreatic cancers, which is correlated with decreased patient survival, indicating its potential roles in malignant development and progression in clinic [9, 10, 12, 13, 14, 15]. In addition, treatment of mice with the inflammation stimulus dextran sulfate sodium (DSS) preferably stimulates p38γ phosphorylation (as compared to p38α) in intestinal epithelial tissues/cells [16], whereas p38α (to a less extent for p38δ) is predominantly activated by lipopolysaccharide (LPS) [17] and tumor necrosis factor (TNF) [18]. In patients with chronic inflammation (arthritis), however, p38α and p38γ, but not other p38s, are both activated [19]. A distinct activation-pattern of p38 family proteins by different stimuli may play an important role in their different biological outcomes and an elevated p38γ RNA/protein in Ras-transformed cells and in cancers indicates its potential as a sustainable therapeutic target for pharmacological intervention.

p38 family MAPK proteins also differently activate their downstream substrates such as kinases and transcription factors [2, 4]. Several kinases, including p38 regulated/activated kinase (PRAK), and mitogen-activated protein kinase–interacting kinase 1 (MNK1), are phosphorylated by p38α and/or p38β in vitro and in cells, but not by other p38 isoforms, whereas MAP kinase-activated protein kinase 2 (MK2) is activated by all p38 family proteins [1, 4]. Transcription factors myocyte enhancer factor 2C (MEF2C) and activating transcription factor-2 (ATF2) are activated by all p38 family proteins [3, 4]. Although c-Jun is activated by p38α and p38γ, this occurs via distinct mechanism: c-Jun is activated by p38α through phosphorylation of the AP-1 partner proteins Sap-1α and ATF2 [1] but activated by p38γ via AP-1-dependent transcription [20, 21, 22]. The different effects of p38 family proteins on downstream kinases and transcription factors may play an important role in their isoform-specific and cell/tissue-dependent activities.

p38γ protein has a unique structure among p38 family proteins, which may determine its capacity to phosphorylate a specific substrate and to signal via a specific pathway through interacting with different proteins [1, 15, 23, 24]. Specifically, p38γ C-terminal contains a PDZ-binding motif that interacts with PDZ-domain containing proteins including its substrate SAP90 [25] and its phosphatase protein tyrosine phosphatase H1 (PTPH1) [11, 15]. Moreover, PDZ motif is required for p38γ to interact with c-Jun in cells [20], which may be important for p38γ to activate AP-1-dependent gene transcription, including c-Jun, matrix metalloproteinase (MMP9) [20], Nanog [21], and epidermal growth factor receptor (EGFR) [22]. Furthermore, p38γ depends on PDZ motif to bind, phosphorylate and activate PTPH1 [26], which is important for PTPH1 to catalyze EGFR dephosphorylation and to promote KRAS-dependent growth [22, 27]. In addition, p38γ binds and/or phosphorylates several proliferative proteins, including DNA topoisomerase IIα (Topo IIα) and estrogen receptor α (ER) in breast cancer [8, 9], heat shock protein 90 (Hsp90) and β-catenin in colon cancer [13, 16], and glucose transporter 2 (Glut2) and phosphofructokinase-2/fructose-2,6-bisphosphatase 3 (PFKFB3) in pancreatic cancer [12]. It is not known, however, if PDZ binding is directly and indirectly involved in p38γ interacting with this group of proliferative proteins. These results together indicate that p38γ may execute its oncogenic activity through interaction with other proliferative proteins dependent and independent of PDZ binding [28].

2. Effects of p38α/β knockout on inflammation and inflammation-associated oncogenesis

Cell culture studies showed p38α inhibits Ras proliferative activity in NIH3T3 fibroblasts by negative feedback in which transient transfection of oncogenic Ras (HRASG61L) stimulates phosphorylation of each member of the co-transfected p38α pathway MKK6 (MAPK kinase 6), p38α, and PRAK (p38-related/activated protein kinase)/MAPK-activated protein kinase 2 (MK2), which in turn suppresses Ras proliferative response [6]. The p38α suppressive activity on Ras oncogene was further demonstrated pharmacologically in intestinal epithelial cells (IEC) in which Ras-dependent soft-agar growth was increased by treatment with the p38α/β inhibitor SB203586 [29]. Moreover, the p38 upstream activator MKK6 and down-stream kinase PRAK and MK2 were further shown to suppress Ras proliferative activity and/or Ras-induced transformation in different in vitro and in vivo systems [6, 30, 31, 32, 33, 34, 35], although recent MK2 knockout studies showed its promoting role in colitis-associated cancer [36]. These results together indicate that the p38α pathway activities in target cells (fibroblasts and epithelial cells) are inhibitory to Ras proliferative activity and oncogenic transformation in cell culture [7] (Table 1, Ref. [12, 16, 34, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64]).

Table 1.Effects of knockout of p38 MAPKs on inflammation and inflammation-associated oncogenesis.
p38 knockout (KO) Major phenotype Reference
-/- (inducible KO) increased lung tumorigenesis [37]
(KRAS)
-/- (KO in MEFs) increased transformation by [38]
Ras and other oncogenes
-/- (inducible KO in MSCs) increased xenograft growth co-injected with [39]
p38α-deleted mesenchymal stromal cells
(MSCs)
liver-specific KO increased liver tumorigenesis [40]
(Den-induced)
IEC-specific KO increased colitis (DSS) [41]
myeloid-specific KO decreased colitis (DSS)
myeloid-specific KO decreased colitis-associated tumorigenesis [42]
(AOM/DSS)
IEC-specific KO increased colitis and colitis-associated cancer [43]
(CAC) (AOM/DSS)
IEC-specific KO* biphasic; increased colon tumorigenesis early [44]
and decreased tumor growth later
Alveolar epithelial type II* biphasic, increased tumorigenesis early [45]
(AE II-specific KO) and decreased tumor formation later (KRAS)
myeloid-specific KO decreased skin inflammation to SDS [46]
increase skin inflammation to UVB
increased skin inflammation to TPA
keratinocyte specific KO decreased skin inflammation to UVB [45]
DC specific KO no effect on skin inflammation to UVB
keratinocyte specific KO increased skin inflammation to LPS/TPA [48]
myeloid specific KO increased skin inflammation to LPS/TPA
DC specific KO decreased skin inflammation to LPS/TPA
T cell specific KO decreased skin inflammation to LPS/TPA
Hepatic-specific KO increased liver tumorigenesis [49]
(Den-induced)
T cell specific KO increased adoptive immunotherapy [50]
(CRISPR-Cas9) increased anti-tumor activity of T cells
-/- (inducible) & decreased lung metastatic of melanoma [51]
Fibroblasts-specific KO decreased lung metastatic of melanoma
Fibroblast-specific KO decreased lung tumorigenesis (KRAS model) [52]
DC-specific KO decreased TH-17 cell differentiation and [53]
decreased IL-23/IL-6 expression
DC-specific KO decreased colitis/colon tumors (AOM/DSS), [54]
increased JNK, IL-10, IFN-γ and
decreased IL-6, TNF, IL-1β, and IL-17
Macrophage specific KO decreased LPS-induced TNFα, IL-12 and IL-18 [55]
Macrophage specific KO decreased colitis in IL-10-/- mice [56]
Keratinocyte-specific KO decreased skin inflammation (to UVB)
MKK3/6-/- in CD4T cells decreased IL-17 [57]
p38α dn Tg in CD4T cells decreased IL-17
MKK3/6-/- in MEFs increased xenograft in immortalized cells [34]
p38β No studies reported
p38γ
-/- decreased TNFα, IL-β and IL-10 [58]
in response to LPS
-/-** decreased colon tumorigenesis [59]
when combined with p38δ-/- (AOM/DSS)
-/-** decreased skin tumorigenesis [60]
when combined with p38δ-/- (DMBA/TPA)
-/- slowed T-cell differentiation [61]
(p38γ-/- more affects in CD4+/CD8+ cells)
(p38δ-/- more affects in CD4-/CD8- cells)
IEC-specific KO decreased colitis and CAC [16]
(AOM/DSS model)
Hepatic-specific KO Decreased liver tumorigenesis [62]
(Den model)
pancreas-specific KO Decreased pancreatic tumorigenesis [12]
(KPC model)
p38δ
-/- decreased skin tumorigenesis [63]
(DMBA model)
-/-** decreased colon tumorigenesis [59]
When combined with p38γ-/-
(AOM/DSS model)
-/-** decreased skin tumorigenesis [60]
when combined with p38γ-/- (DMBA/TPA)
-/- decreased mammary tumorigenesis [64]
(PyMT model)
The global knockout is indicated by a sign “-/-”, whereas conditional knockout (KO) is shown as cell/tissue-specific KO through the Cre recombinase technology. Tumors were induced by transgenic expression of the indicated oncogene and/or by treatment of mice with the indicated carcinogen ± inflammation stimuli (please see details in the indicated references). *indicates a biphasic effect with enhanced tumorigenesis by inducible p38α conditional KO during tumor initiation and with decreased tumor growth and/or metastasis after tumor established, and **shows similar phenotypes in p38γ KO and p38δ KO mice, which is more substantial in their double KO mice. Effects of p38γ in experimental and clinic cancer were recently reviewed [74] and results from this table are summarized in Table 2.

Systemic effects of p38α in inflammation and in inflammation-associated oncogenesis have been investigated by knockout (KO) studies in mice. Because global p38α KO is embryonic lethal [65, 66], inducible and/or conditional p38α KO was developed. Specific p38α KO in macrophages leads to changes in pro-inflammatory cytokines TNF, IL-6, and anti-inflammatory cytokine IL-10 in bone marrow-derived macrophages (BMDM) in a manner dependent of stimuli and of treatment time, which is blocked by IL-10 antibody, indicating a proinflammatory response [56]. Further, myeloid p38α KO decreases colitis, inhibits colitis-associated cancer (CAC) [42], and prolongs survival of IL-10-/- mice, indicating that myeloid p38α is pro-inflammatory and oncogenic [56]. A pro-inflammatory role of p38α is also demonstrated by a decrease in 2,4-dinitrofluorobenzene (DNFB)-induced ear swelling in mice with p38α KO in dendritic cells (DCs) and in T cells, although myeloid-specific p38α KO had an opposite effect [48]. Moreover, p38α KO in DCs inhibits dextran sodium sulfate (DSS)-induced colitis and attenuates DSS/azoxymethane (AOM)-induced CAC in association with decreased neutrophil infiltration and with changes in multiple cytokines in colon tissues [54], further indicating the pro-inflammatory and oncogenic role of p38α in immune cells (Fig. 1). This conclusion is further supported by decreased lethality in mice after the treatment with lipopolysaccharide (LPS) in which p38α is specifically deletion in macrophage in association with reduced blood levels of pro-inflammatory cytokines TNF, IL-12, and IL-18 [55]. Moreover, there is attenuated colitis and decreased inflammatory cytokine expression (after DSS) in mice with myeloid-specific p38α KO [41]. Myeloid p38α is also important for DSS-induced skin inflammation [46] and p38α KO in DCs, but not in macrophages or T cells, inhibits TH17 differentiation, decreases IL-17 levels, and suppresses autoimmune inflammation [67]. In addition, inhibition of p38α activity by expressing a dominant negative (dn) mutant in CD4 T cells decreases IL-17 expression and reduces the severity of allergic encephalomyelitis (EAE) [57]. Studies with a CRISPR-Cas9 screening of primary T cells further showed that p38α deletion increases the efficacy of mouse anti-tumor T cells [50, 68], thus demonstrating an oncogenic role of p38α in T cells. A recent study further showed that p38α activity (the phospho-p38α/total p38α ratio) in leukocytes isolated from the patient peripheral blood with metastatic melanoma is increased as compared to those without metastasis, and predicts decreased patient survival, and that p38α KO specifically in fibroblasts attenuates lung metastasis of melanoma in mice [51]. Moreover, specific deletion of p38α from fibroblasts also inhibits KRAS-induced lung tumorigenesis [52]. These results together indicate that p38α activity in stromal cells (immune cells and fibroblasts) overall is pro-inflammatory and/or oncogenic [48, 54, 67] (Fig. 1) (Tables 1,2).

Fig. 1.

p38 (α, β, γ, and δ) family proteins regulate inflammation and inflammation-associated oncogenesis by isoform-specific and cell/tissue-dependent mechanism. p38α activity in immune cells is mostly proinflammatory and oncogenic while in epithelial cells (and other target cells such as MEFs) is anti-inflammatory and tumor suppressive. p38γ and p38δ activity in immune and epithelial cells is both proinflammatory and oncogenic. * Biphasic effects of inducible p38α KO in intestinal epithelial cells and in lung epithelial projector cells, i.e., p38α is a tumor suppressor in cancer initiation stage but is oncogenic in advanced stage likely via tumor-stromal interactions. No p38β studies on inflammation and/or cancer have been reported. ** Only studies of conditional p38δ and combined p38γ/p38δ KO in myeloid cells were reported.

Table 2.Summary of p38 MAPKs in inflammation and cancer.
Pro-inflammatory Anti-inflammatory Tumor-suppressive Oncogenic Oncogenic Others
Knockout Epithelial Immune cells Epithelial Immune cells Epithelial Immune cells Epithelial Immune cells Fibroblast
p38α X* X X* X X X
p38β ?
p38γ X X X
p38δ X X X
*Response differs in a stimulus- and cell/tissue-specific manner.

Studies with specific p38α KO in epithelial cells in which tumor develops, however, showed that p38α is anti-inflammatory with a tumor suppressor activity [41, 40, 49, 43, 44]. Experiments in mice with intestinal epithelial cell (IEC)-specific p38α KO, for example, showed increased IEC proliferation, enhanced colitis severity and/or colon tumorigenesis after the treatment with DSS ± azoxymethane (AOM) as compared to control mice [41, 43, 44]. An increase in the carcinogen diethyl nitrosamine (DEN)-induced liver tumorigenesis was also observed in mice with hepatic-specific p38α KO [40, 49]. Moreover, studies in H-Ras-transformed or immortalized fibroblasts showed increased in vivo xenograft formation of mouse embryonic fibroblasts (MEFs) lacking p38α [38] and its activator MKK3 and MKK6 [34]. Moreover, experiments with inducible p38α global knockout revealed that p38α KO increases lung stem cell proliferation and KRAS-induced lung tumorigenesis [37]. In addition, co-injection of p38α-deleted mesenchymal stem cells (MSCs) increases xenograft growth of human colon cancer cells in nude mice in association with enhanced angiogenesis [39]. However, inhibition of p38α nuclear translocation by a peptide attenuates AOM/DSS-induced colon cancer, likely through targeting p38α in multiple cell-types and tissues [69]. These results together indicate that p38α activity in target cells (epithelial, fibroblasts) and in co-injected MSCs is anti-inflammatory and/or tumor-suppressive in response to carcinogen, inflammation stimulus and/or RAS oncogene (Fig. 1).

Recent studies further showed that inducible p38α KO at a late stage in intestinal epithelial cells (65 days after AOM/DSS administration to induce colon tumor) and in alveolar epithelial progenitor cells (20 weeks after induction of KRASG12V expression in lungs) decreases tumorigenesis, despite the initial increase in tumorigenesis in both tissues [44, 45]. Mechanisms involved however are mostly unclear and may involve epithelial p38α signaling interaction with stromal once tumor reaches a certain size [52, 70]. This speculation is supported by the fact that p38α silencing in pancreatic cancer cells inhibits the cell growth in vitro but increases the xenograft formation of the same cells in mice [71] and that p38α in fibroblasts promotes lung metastasis of melanoma [51] and lung tumorigenesis [52]. These results indicate a stage-specific role of epithelial p38α in tumorigenesis and metastasis likely through signaling interactions with stromal tissues. Although studies also showed a distinct role of p38α vs p38β in cell survival and cell death [71, 72], p38β is generally believed to be redundant and its global KO did not show major phenotypes [73]. These results together indicate that p38α in epithelial cells has a dual role in oncogenesis, i.e., anti-inflammatory as a tumor suppressor at the tumor initiation but oncogenic once tumor is established or becomes metastatic (Fig. 1) (Tables 1,2).

3. Effects of p38γ/δ knockout on inflammation and inflammation-associated oncogenesis

Genetic studies showed that mice with global p38γ and/or p38δ knockout are phenotypically normal, which however results in a decrease in multiple cytokines in response to lipopolysaccharide [LPS, a toll-like receptor 4 (TLR4) ligand] in bone marrow-derived macrophages (BMDM) [58, 75]. Although global p38γ knockout alone has no significant effect on 7,12-dimethylbenz(a)anthracene (DMBA)/tetradecanoyl-phorbol-13-acetate (TPA)-induced skin tumorigenesis as compared with wild-type (WT) mice, there is attenuated tumorigenesis in p38δ KO mice with a more substantial effect in mice with its combined KO with p38γ [60]. In colon cancer studies, p38γ and p38δ global KO has no major impact on chronic inflammation but decreases acute inflammation in intestine tissues in response to DSS [59]. Moreover, mice with myeloid-specific p38γ and/or p38δ KO are resistant to diet-induced fatty liver, hepatic triglyceride, and glucose intolerance in association with defective migration of neutrophils to the damaged liver [76]. Analyses of global p38γ and/or p38δ KO mice further showed that p38δ and p38γ KO differentially regulates T cell differentiation at different stages as compared with WT mice [61]. Separate studies showed that both myeloid-specific and global p38δ KO decreased alveolar neutrophil accumulation and attenuated acute lung injury [77], whereas combined p38γ/p38δ myeloid-specific and global KO protects mice against fungal infection and inhibits leukocyte recruitment to infected kidneys [78]. These results together indicate that systemic p38γ and p38δ activity and their signaling in immune cells (only KO data available in myeloid cells) are mostly pro-inflammatory and/or oncogenic (Fig. 1).

Recent genetic studies in mouse cancer models further showed that systemic and epithelial p38γ in gastrointestinal (GI) system is essential for tumorigenesis. Global p38γ and p38δ KO attenuates colitis-associated cancer (CAC) with their combined KO having more significant effects than either alone, indicating a cooperative oncogenic activity of systemic p38γ and p38δ [59]. Moreover, IEC-specific p38γ KO alone decreases pro-inflammatory cytokines (IL-6, IL-1β and TNF), inhibits the β-catenin/Wnt pathway in colonic tissues, and attenuates DSS-induced colitis and AOM/DSS-induced CAC [16]. Importantly, oral application of a p38γ selective pharmacological inhibitor pirfenidone (PFD) [79, 80] depends on epithelial p38γ to decrease p38γ phosphorylating its substrates and to reduce cytokine’s levels in tumor tissues, and to inhibit tumorigenesis, suggesting a novel strategy to block colon tumorigenesis by targeting epithelial p38γ [16]. p38γ was further shown to phosphorylate RB and to drive cell cycle progression, and hepatic p38γ KO and systemic application of PFD both block diethyl nitrosamine (DEN)-induced liver tumorigenesis [62]. Our recent studies further showed that p38γ mediates KRAS oncogene signaling to activate the glycolytic pathway in pancreatic ductal cancer cells (Pdac) and that specific p38γ KO in pancreatic epithelial cells inhibits pancreatitis, reduces cytokine levels, and decreases pancreatic tumorigenesis in KPC mice [12]. Moreover, epithelial p38γ is required for PFD to suppress glycolytic pathways, to block pancreatic tumorigenesis in KPC mice, and to inhibit Pdac xenograft growth [12]. Together, these results demonstrate that epithelial p38γ is essential for colon, liver and pancreatic tumorigenesis and its pharmacological inhibitor PFD may have therapeutic potentials to block their development, growth, and progression (Fig. 1) (Tables 1,2).

Studies also showed that p38δ is required for tumorigenesis in certain tissues. An early study showed that global p38δ KO blocks DMBA/TPA-induced skin tumorigenesis [63]. Studies from Cuenda lab further showed that global p38δ KO alters expression of several cytokines in response to DSS [59]. Although combined global p38γ/p38δ KO appears to achieve more substantial effects in regulating cytokines and in inhibiting CAC than either alone in DSS/AOM mouse model, analyses of chimeric mice of WT with p38γ/p38δ-/- animals revealed a critical role of hematopoietic, but not epithelial, p38γ/p38δ in regulation of inflammatory mediators and immune cell recruitment [59]. A protective effect of global p38δ KO on DMBA/TPA-induced skin tumorigenesis was observed in association with decreased cytokines and chemokines in skin tissues, which are further enhanced in p38γ/δ double KO mice [60]. A recent study further showed that conditional knockout of p38δ in mammary epithelial cells decreases the viral oncogene PyMT-induced breast tumorigenesis in mice [64]. These results together indicate that systemic and epithelial p38δ, as in the case with p38γ, is pro-inflammatory and oncogenic (Fig. 1) (Tables 1,2).

4. Implications of cell/tissue-type dependent and isoform-specific effects of p38 MAPKs in inflammation and in inflammation-associated oncogenesis

Mechanisms for cell/tissue-dependent and isoform-specific roles of p38 family proteins in inflammation and inflammation-associated oncogenesis are largely unknown. Although different p38 MAPK isoforms may regulate different sets of inflammation mediators and/or different groups of downstream molecules in response to different stimuli and/or in different cells/tissues, there is still a lack of experimental evidence to support this hypothesis. While it is difficult to systemically compare intrinsic activities of p38 family proteins in immune cells due to lack of genetic evidence, p38α and p38γ in epithelial cells appear to be antagonistic. This effect has been observed at the level of protein, cell, and disease. At protein level, for example, p38α and p38γ both phosphorylate the tumor suppressor Rb at different sites leading to an opposite effect on cell-cycle progression. Specifically, p38γ phosphorylates Rb at S807/S811 and stimulates G1/S transition [62], whereas p38α phosphorylates Rb at S429/T252 and slows cell-cycle progression [81]. Although Rb phosphorylation at these different sites is not known to be sufficient to trigger the opposite effect on cell-cycle progression, this mechanism may contribute to the tumor suppressor activity of p38α and oncogenic activity of p38γ. At cellular level, we showed an antagonizing effect of p38α and p38γ in stress response and in KRAS transformation in which p38α transfection directly depletes cellular p38γ protein by a ubiquitination-dependent mechanism [82] and that inhibition of p38α activity with SB203580 increases p38γ protein levels [20]. At disease level, increased p-p38α in pancreatic cancer tissues couples with increased patient’s survival, indicating its tumor-suppressive activity [83], whereas upregulated p38γ in the same cancer predicts decreased patient survival, suggesting its oncogenic effect [12]. Thus, p38γ and p38α can antagonize each other toward a protein substrate in stress or oncogene-induced cellular outcome and in clinical cancer development and progression. This cross-restrained activity of p38α and p38γ could complicate therapeutic gain when their isoform-specific pharmacological inhibitors are used in systemic intervention. Please see recent outstanding reviews about p38 MAPKs and inhibitors [2, 84].

Cell/tissue-specific effects of p38 family proteins will also have important implications for using their pharmacological inhibitors to regulate inflammation and inflammation-driven oncogenesis systemically. Although p38α in immune cells is pro-inflammatory, application of its inhibitor SB203580 does not improve clinical symptoms of DSS-induced colitis in mice [41]. This might occur as an integration of its inhibition of pro-inflammatory p38α activity in immune system and of its blockade of anti-inflammatory effect of p38α in intestinal epithelial cells (Fig. 1) [41]. These experimental results are consistent with a poor outcome of clinical trials using an oral p38α inhibitor BIRB in the treatment of Crohn’ disease [85]. On the other hand, p38γ activity in immune cells and in epithelial cells is both pro-inflammatory and oncogenic (Fig. 1) and its inhibitor PFD therefore showed a significant and consistent inhibitory effect on inflammation and inflammation-associated oncogenesis as observed in mouse models of colon, liver, and pancreatic cancer [12, 16, 62]. Considering of cell/tissue-dependent and isoform-specific effects of p38 family proteins is therefore critical for development of effective small molecular p38 inhibitors against inflammation and inflammation-driven cancer in therapeutic intervention.

Abbreviations

AOM, azoxymethane; ATF2, activating transcription factor-2; BMDM, bone marrow-derived macrophages; DMBA, 12-dimethylbenz(a)anthracene; DCs, dendritic cells; DNFB, 2,4-dinitrofluorobenzene; DSS, dextran sulfate sodium; EGFR, epidermal growth factor receptor; ER, estrogen receptor α; Hsp90, heat shock protein 90 alpha; IEC, intestinal epithelial cell; “-/-”, global deletion; KO, knockout; MAPKs, mitogen-activated protein kinases; MEFs, mouse embryonic fibroblasts; MEF2C, myocyte enhancer factor 2C; MKK3 or 6, MAPK kinase 3 and/or 6; MMP9, matrix metalloproteinase; LPS, lipopolysaccharide; KPC, LSL-KrasG12D/+: LSL-Trp53R172H/+: Pdx1-Cre mice; MAPK, mitogen-activated protein kinase; p38α MAPK, MAPK14; p38β, MAPK11; p38γ, MAPK12; p38δ, MAPK13; PRAK, p38 regulated/activated kinase; MK2, MAP kinase-activated protein kinase 2; MNK1, mitogen-activated protein kinase-interacting kinase 1; PFKFB3, phosphofructokinase-2/fructose-2,6-bisphosphatase 3; PTPH1, protein tyrosine phosphatase H1; PyMT, polyomavirus middle T antigen; TPA, 12-tetradecanoylphorbol-13-acetate; WT, wild-type.

Author contributions

JZQ, GC—concept development and manuscript writing; HX, XMQ—discussion of the manuscript and figure preparation.

Ethics approval and consent to participate

Not applicable.

Acknowledgment

We are very grateful to former Chen’s lab members for their contributions to the research work.

Funding

This work has been supported by grants from NIH (R01 CA245977), VA (I01 BX005066), DOD (BC141898) and MCW Cancer Center (to GC).

Conflict of interest

The authors declare no conflict of interest. GC is serving as one of the Editorial Board members of this journal. We declare that GC had no involvement in the peer review of this article and has no access to information regarding its peer review. Full responsibility for the editorial process for this article was delegated to GP.

References
[1]
Ono K, Han J. The p38 signal transduction pathway Activation and function. Cellular Signaling. 2000; 12: 1–13.
[2]
Canovas B, Nebreda AR. Diversity and versatility of p38 kinase signaling in health and disease. Nature Reviews Molecular Cell Biology. 2021; 22: 346–366.
[3]
Cuenda A, Sanz-Ezquerro J. p38γ and p38δ: from spectators to key physiological players. Trends in Biochemical Sciences. 2017; 42: 431–42.
[4]
Han J, Wu J, Silke J. An overview of mammalian p38 mitogen-activated protein kinases, central regulators of cell stress and receptor signaling. F1000 Faculty Reviews. 2020; 9: 653.
[5]
Cuenda A, Rousseau S. P38 MAP-Kinases pathway regulation, function and role in human diseases. Biochimica Biophysica Acta (BBA) - Molecular Cell Research. 2007; 1773: 1358–1375.
[6]
Chen G, Hitomi M, Han J, Stacey DW. The p38 pathway provides negative feedback to Ras proliferative signaling. Journal of Biological Chemistry. 2000; 275: 38973–80.
[7]
Loesch M. The p38 MAPK stress pathway as a tumor suppressor or more? Frontiers in Bioscience. 2008; 13: 3581–3593.
[8]
Qi X, Hou S, Lepp A, Li R, Basir Z, Lou Z, et al. Phosphorylation and Stabilization of Topoisomerase IIα Protein by p38γ Mitogen-activated Protein Kinase Sensitize Breast Cancer Cells to its Poisons. Journal of Biological Chemistry. 2011; 286: 35883–35890.
[9]
Qi X, Zhi H, Lepp A, Wang P, Huang J, Basir Z, et al. P38γ Mitogen-activated Protein Kinase (MAPK) Confers Breast Cancer Hormone Sensitivity by Switching Estrogen Receptor (ER) Signaling from Classical to Nonclassical Pathway via Stimulating ER Phosphorylation and c-Jun Transcription. Journal of Biological Chemistry. 2012; 287: 14681–14691.
[10]
Tang J, Qi X, Mercola D, Han J, Chen G. Essential Role of p38γ in K-Ras Transformation Independent of Phosphorylation. Journal of Biological Chemistry. 2005; 280: 23910–23917.
[11]
Qi X, Wang F, Chen G. P38 Gamma MAPK. Encyclopedia of Signaling Molecules. 2018; 10: 3718–3727.
[12]
Wang F, Qi X, Wertz R, Mortensen M, Hagen C, Evans J, et al. P38γ MAPK is Essential for Aerobic Glycolysis and Pancreatic Tumorigenesis. Cancer Research. 2020; 80: 3251–3264.
[13]
Qi X, Xie C, Hou S, Li G, Yin N, Dong L, et al. Identification of a ternary protein-complex as a therapeutic target for K-Ras-dependent colon cancer. Oncotarget. 2014; 5: 4269–4282.
[14]
Rosenthal DT, Lyer H, Escudero S, Bao L, Wu Z, Ventura AC, et al. p38γ promotes breast cancer motility and metastasis through regulation of RhoC GTPase, cytoskeletal architecture, and a novel leading edge behavior. Cancer Research. 2011; 71 6338–49.
[15]
Hou SW, Zhi H, Pohl N, Loesch M, Qi X, Li R, et al. PTPH1 dephosphorylates and cooperates with p38γ MAPK to increases Ras oncogenesis through PDZ-mediated interaction. Cancer Research. 2010; 70: 2901–10.
[16]
Yin N, Qi X, Tsai S, Lu Y, Basir Z, Oshima K, et al. P38γ MAPK is required for inflammation-associated colon tumorigenesis. Oncogene. 2016; 35: 1039–1048.
[17]
Hale KK, Trollinger D, Rihanek M, Manthey CL. Differential expression and activation of p38 mitogen-activated protein kinase alpha, beta, gamma, and delta in inflammatory cell lineages. Journal of Immunology. 1999; 162: 4246–52.
[18]
Mukaro V, Quach A, Gahan ME, Boog B, Huang ZH, Gao Xea. Small tumor necrosis factor receptor biologics inhibit the tumor necrosis factor-p38 signaling axis and inflammation. Nature Communications. 2018; 9: 1635.
[19]
Korb A, Tohidast-Akrad M, Cetin E, Axmann R, Smolen J, Schett G. Differential tissue expression and activation of p38 MAPK α, β, γ, and δ isoforms in rheumatoid arthritis. Arthritis & Rheumatism. 2006; 54: 2745–2756.
[20]
Loesch M, Zhi H, Hou S, Qi X, Li R, Basir Z, et al. P38γ MAPK Cooperates with c-Jun in trans-Activating Matrix Metalloproteinase 9. Journal of Biological Chemistry. 2010; 285: 15149–15158.
[21]
Qi X, Yin N, Ma S, Lepp A, Tang J, Jing W, et al. P38γ MAPK is a Therapeutic Target for Triple‐Negative Breast Cancer by Stimulation of Cancer Stem‐Like Cell Expansion. STEM CELLS. 2015; 33: 2738–2747.
[22]
Yin N, Lepp A, Ji Y, Mortensen M, Hou S, Qi X, et al. The K-Ras effector p38γ MAPK confers intrinsic resistance to tyrosine kinase inhibitors by stimulating EGFR transcription and EGFR dephosphorylation. Journal of Biological Chemistry. 2017; 292: 15070–15079.
[23]
Remy G, Risco AM, Iñesta-Vaquera FA, González-Terán B, Sabio G, Davis RJ, et al. Differential activation of p38MAPK isoforms by MKK6 and MKK3. Cellular Signalling. 2010; 22: 660–667.
[24]
Hou S, Lepp A, Chen G. p38 gamma MAP kinase UCSD-Nature Molecular Pages. 2010; doi:1038/mp.a001720.01.
[25]
Sabio G, Reuver S, Feijoo C, Hasegawa M, Thomas GM, Centeno F, et al. Stress- and mitogen-induced phosphorylation of the synapse-associated protein SAP90/PSD-95 by activation of SAPK3/p38γ and ERK1/ERK2. Biochemical Journal. 2004; 380: 19–30.
[26]
Hou S, Padmanaban S, Qi X, Leep A, Mirza S, Chen G. p38γ MAPK signals through phosphorylating its phosphatase PTPH1 in regulating Ras oncogenesis and stress response. Journal of Biological Chemistry. 2012; 287: 27895–27905.
[27]
Ma S, Yin N, Qi X, Pfister SL, Zhang M, Ma R, et al. Tyrosine dephosphorylation enhances the therapeutic target activity of epidermal growth factor receptor (EGFR) by disrupting its interaction with estrogen receptor (ER) Oncotarget. 2015; 6: 13320–13333.
[28]
Qi X, Wang F, Mortensen M, Wertz R, Chen G. Targeting an oncogenic kinase/phosphatase signaling network for cancer therapy. Acta Pharmaceutica Sinica B. 2018; 8: 511–517.
[29]
Pruitt K, Pruitt WM, Bilter GK, Westwick JK, Der CJ. Raf-independent Deregulation of p38 and JNK Mitogen-activated Protein Kinases are Critical for Ras Transformation. Journal of Biological Chemistry. 2002; 277: 31808–31817.
[30]
Awad MM, Enslen H, Boylan JM, Davis RJ, Gruppuso PA. Growth Regulation via p38 Mitogen-activated Protein Kinase in Developing Liver. Journal of Biological Chemistry. 2000; 275: 38716–38721.
[31]
Sun P, Yoshizuka N, New L, Moser BA, Li Y, Liao R, et al. PRAK is Essential for ras-Induced Senescence and Tumor Suppression. Cell. 2007; 128: 295–308.
[32]
Zhang R, Murakami S, Coustry F, Wang Y, de Crombrugghe B. Constitutive activation of MKK6 in chondrocytes of transgenic mice inhibits proliferation and delays endochondral bone formation. Proceedings of the National Academy of Sciences. 2007; 103: 365–370.
[33]
Wuestefeld T, Pesic M, Rudalska R, Dauch D, Longerich T, Kang T, et al. A Direct in Vivo RNAi Screen Identifies MKK4 as a Key Regulator of Liver Regeneration. Cell. 2013; 153: 389–401.
[34]
Brancho D, Tanaka N, Jaeschke A, Ventura J, Kelkar N, Tanaka Y, et al. Mechanism of p38 MAP kinase activation in vivo. Genes & Development. 2003; 17: 1969–1978.
[35]
Kobayashi Y, Qi X, Chen G. MK2 Regulates Ras Oncogenesis through Stimulating ROS Production. Genes & Cancer. 2012; 3: 521–530.
[36]
Suarez-Lopez L, Sriram G, Kong YW, Morandell S, Merrick KA, Hernandez Y, et al. MK2 contributes to tumor progression by promoting M2 macrophage polarization and tumor angiogenesis. Proceedings of the National Academy of Sciences. 2018; 115: E4236–E4244.
[37]
Ventura JJ, Tenbaum S, Perdiguero E, Huth M, Guerra C, Barbacid M, et al. p38α MAP kinase is essential in lung stem and progenictor cell proliferation and differentiation. Nature Genetics. 2007; 39: 750–758.
[38]
Dolado I, Swat A, Ajenjo N, De Vita G, Cuadrado A, Nebreda AR. P38α MAP Kinase as a Sensor of Reactive Oxygen Species in Tumorigenesis. Cancer Cell. 2007; 11: 191–205.
[39]
Batlle R, Andrés E, Gonzalez L, Llonch E, Igea A, Gutierrez-Prat N, et al. Regulation of tumor angiogenesis and mesenchymal–endothelial transition by p38α through TGF-β and JNK signaling. Nature Communications. 2019; 10: 3071.
[40]
Hui L, Bakiri L, Mairhorfer A, Schweifer N, Haslinger C, Kenner L, et al. P38α suppresses normal and cancer cell proliferation by antagonizing the JNK–c-Jun pathway. Nature Genetics. 2007; 39: 741–749.
[41]
Otsuka M, Kang YJ, Ren J, Jiang H, Wang Y, Omata M, et al. Distinct Effects of p38α Deletion in Myeloid Lineage and Gut Epithelia in Mouse Models of Inflammatory Bowel Disease. Gastroenterology. 2010; 138: 1255–1265.
[42]
Youssif C, Cubillos-Rojas, Comalade M, LIonch E, Perna C, Djouder Nea. Myeloid p38α signaling promotes intestinal IGF-1 production and inflammation-associated tumorigenesis. EMBO Molecular Medicine. 2018; 10: e8403.
[43]
Wakeman D, Schneider JE, Liu J, Wandu WS, Erwin CR, Guo J, et al. Deletion of p38-alpha mitogen-activated protein kinase within the intestinal epithelium promotes colon tumorigenesis. Surgery. 2012; 152: 286–293.
[44]
Gupta J, Barantes IB, Igea A, Sakellariou S, Pateras I, Gorgoulis VGea. Dual function of p38α MAPK in colon cancer: suppression of colities-associated tumor initiation but requirement for cancer cell survival. Cancer Cell. 2014; 25: 484–550.
[45]
Vitos-Faleato J, Real SM, Gutierrez-Prat N, Villanueva A, Llonch E, Drosten M, et al. Requirement for epithelial p38α in KRAS-driven lung tumor progression. Proceedings of the National Academy of Sciences. 2020; 117: 2588–2596.
[46]
Kim C, Sano Y, Todorova K, Carlson BA, Arpa L, Celada A, et al. The kinase p38α serves cell type-specific inflammatory functions in skin injury and cooridinates pro- and anti-inflammatory gene expression. Nature Immunology. 2008; 9: 1019–1027.
[47]
Sano Y, Park JM. Loss of epidermal p38α signaling prevents UVR-induced inflammation via acute and chronic mechanisms. Journal of Investigative Dermatology. 2014; 134: 2231–40.
[48]
Ritprajak P, Hayakawa M, Sano Y, Otsu K, Park JM. Cell type-specific targeting dissociates the therapeutic from the adverse effects of protein kinase inhibition in allergic skin disease. Proceedings of the National Academy of Sciences. 2012; 109: 9089–9094.
[49]
Sakurai T, Kudo M, Umemura A, He G, Elsharkawy AM, Seki E, et al. p38α inhibits liver fibrogenesis and consequent hepatocarcinogenesis by curtailing accumulation reactive oxygen species. Cancer Research. 2012; 73: 215–224.
[50]
Gurusamy D, Henning AN, Yamamoto TN, Yu Z, Zacharakis N, Krishna S, et al. Multi-phenotype CRISPR-Cas9 Screen Identifies p38 Kinase as a Target for Adoptive Immunotherapies. Cancer Cell. 2020; 37: 818–833.e9.
[51]
Gui J, Zahedi F, Ortiz A, Cho C, Katlinski KV, Alicea-Torres Kea. Activation of p38α stress-activated proein kinase drives the formation of the pre-metastatic niche in the lungs. Nature Cancer. 2020; 1: 603–609.
[52]
Brichkina A, Bertero T, Loh HM, Nguyen NTM, Emelyanov A, Rigade S, et al. P38MAPK builds a hyaluronan cancer niche to drive lung tumorigenesis. Genes & Development. 2016; 30: 2623–2636.
[53]
Huang J, Nguyen-McCarty M, Hexner EO, Danet-Desnoyers G, Klein PS. Maintenance of hematopoietic stem cells through regulation of Wnt and mTOR pathways. Nature Medicine. 2012; 18: 1778–1785.
[54]
Zheng T, Zhang B, Chen C, Ma J, Meng D, Huang J, et al. Protein kinase p38α signaling in dendritic cells regulates colon inflammation and tumorigenesis. Proceedings of the National Academy of Sciences. 2018; 115: E12313–E12322.
[55]
Kang YJ, Chen J, Otsuka. M., Mols J, Ren S, Wang Y, et al. Macrophage deletion of p38α partially impairs lipopolysaccharide-induced cellular activation. Journal of Immunology. 2008; 180: 5075–5082.
[56]
Raza A, Crothers JW, McGill MM, Mawe GM, Teuscher C, Krementsov DN. Anti-inflammatory roles of p38α MAPK in macrophages are context dependent and require IL-10. Journal of Leukocyte Biology. 2017; 102: 1–9.
[57]
Noubade R, Krementsov DN, del Rio R, Thornton T, Nagaleekar V, Saligrama N, et al. Activation of p38 MAPK in CD4 T cells controls IL-17 production and autoimmune encephalomyelitis. Blood. 2011; 118: 3290–3300.
[58]
Risco A, del Fresno C, Mambol A, Alsina-Beauchamp D, MacKenzie KF, Yang H-, et al. P38 and p38 kinases regulate the Toll-like receptor 4 (TLR4)-induced cytokine production by controlling ERK1/2 protein kinase pathway activation. Proceedings of the National Academy of Sciences. 2012; 109: 11200–11205.
[59]
del Reino P, Alsina-Beauchamp D, Escós A, Cerezo-Guisado MI, Risco A, Aparicio N, et al. Pro-Oncogenic Role of Alternative p38 Mitogen-Activated Protein Kinases p38γ and p38δ, Linking Inflammation and Cancer in Colitis-Associated Colon Cancer. Cancer Research. 2014; 74: 6150–6160.
[60]
Zur R, Garcia-Ibanez L, Nunez-Buiza A, Aparicio N, Liappas G, Escós A, et al. Combined deletion of p38γ and p38δ reduces skin inflammation and protects from carcinogenesis. Oncotarget. 2015; 6: 12920–12935.
[61]
Risco A., Martin-Serrano M, Barber DF, Cuenda A. p38γ and p38δ Are Involved in T Lymphocyte Development. Frontiers in Immunology. 2018; 9: 65.
[62]
Tomás-Loba A, Manieri E, González-Terán B, Mora A, Leiva-Vega L, Santamans AM, et al. P38γ is essential for cell cycle progression and liver tumorigenesis. Nature. 2019; 568: 557–560.
[63]
Schindler EM, Hindes A, Gribben EL, Burns CJ, Yin Y, Lin M, et al. P38δ Mitogen-Activated Protein Kinase is Essential for Skin Tumor Development in Mice. Cancer Research. 2009; 69: 4648–4655.
[64]
Wada M, Canals D, Adada M, Coant N, Salama MF, Helke KL, et al. P38 delta MAPK promotes breast cancer progression and lung metastasis by enhancing cell proliferation and cell detachment. Oncogene. 2017; 36: 6649–6657.
[65]
Tamura K, Sudo T, Senftleben U, Dadak AM, Johnson R, Karin M. Requirement for p38α in Erythropoietin Expression. Cell. 2000; 102: 221–231.
[66]
Adams RH, Porras A, Alonso G, Jones M, Vintersten K, Panelli S, et al. Essential Role of p38α MAP Kinase in Placental but not Embryonic Cardiovascular Development. Molecular Cell. 2000; 6: 109–116.
[67]
Huang G, Wang Y, Vogel P, Kanneganti T, Otsu K, Chi H. Signaling via the kinase p38α programs dendritic cells to drive TH17 differentiation and autoimmune inflammation. Nature Immunology. 2012; 13: 152–161.
[68]
Chan JD, Beavis PA, Darcy PK. P38 Kinase: a Key Target for Driving Potent T Cells for Adoptive Immunotherapy. Cancer Cell. 2020; 37: 756–758.
[69]
Maik-Rachline G, Zehoral E, Hanoch T, Bleins J, Seger R. The nuclear translocation of the kinase p38 and JNK promotes inflammation-induced cancer. Science Signaling. 2018; 11: eaao3428.
[70]
Alspach E, Flanagan KC, Luo X, Ruhland MK, Huang H, Pazolli E, et al. P38MAPK Plays a Crucial Role in Stromal-Mediated Tumorigenesis. Cancer Discovery. 2014; 4: 716–729.
[71]
TIAN X, TRAUB B, XIE X, ZHOU S, HENNE-BRUNS D, KNIPPSCHILD U, et al. Opposing Oncogenic Functions of p38 Mitogen-activated Protein Kinase Alpha and Beta in Human Pancreatic Cancer Cells. Anticancer Research. 2020; 40: 5545–5556.
[72]
Ferrari G, Terushkin V, Wolff MJ, Zhang X, Valacca C, Poggio Pea. TGF-β1 endothelial cell apoptosis by shifting VEGF activation of p38MAPK from the prosurvival p38β to proapoptotic p38α. Molecular Cancer Research. 2012; 10: 605–14.
[73]
Beardmore VA, Hinton HJ, Eftychi C, Apostolaki M, Armaka M, Darragh J, et al. Generation and Characterization of p38β (MAPK11) Gene-Targeted Mice. Molecular and Cellular Biology. 2005; 25: 10454–10464.
[74]
Xu W, Liu R, Dai Y, Hong S, Dong H, Wang H. The Role of p38γ in Cancer: from review to outlook. International Journal of Biological Sciences. 2021; 17: 4036–4046.
[75]
Sabio G, Arthur JSC, Kuma Y, Peggie M, Carr J, Murray-Tait V, et al. P38γ regulates the localisation of SAP97 in the cytoskeleton by modulating its interaction with GKAP. the EMBO Journal. 2005; 24: 1134–1145.
[76]
González‐Terán B, Matesanz N, Nikolic I, Verdugo MA, Sreeramkumar V, Hernández‐Cosido L, et al. P38γ and p38δ reprogram liver metabolism by modulating neutrophil infiltration. the EMBO Journal. 2016; 35: 536–552.
[77]
Ittner A, Block H, Reichel CA, Varjosalo M, Gehart H, Sumara G, et al. Regulation of PTEN activity by p38δ-PKD1 signaling in neutrophils confers inflammatory responses in the lung. Journal of Experimental Medicine. 2012; 209: 2229–2246.
[78]
Alsina‐Beauchamp D, Escós A, Fajardo P, González‐Romero D, Díaz‐Mora E, Risco A, et al. Myeloid cell deficiency of p38γ/p38δ protects against candidiasis and regulates antifungal immunity. EMBO Molecular Medicine. 2018; 10.
[79]
Ozes O, Blatt LM, Seiwert SD. Use of pirfenidone in therapeutic regimens. United States Patent-US 7,407,973 B2 2008; Aug. 5th:1–46.
[80]
Richeldi L, Yasothan U, Kirkpatrick P. Pirfenidone. Nature Reviews Drug Discovery. 2011; 10: 489–490.
[81]
Gubern A, Joaquin M, Marquès M, Maseres P, Garcia-Garcia J, Amat R, et al. The N-Terminal Phosphorylation of RB by p38 Bypasses its Inactivation by CDKs and Prevents Proliferation in Cancer Cells. Molecular Cell. 2016; 64: 25–36.
[82]
Qi X, Pohl NM, Loesch M, Hou S, Li R, Qin J, et al. P38α Antagonizes p38γ Activity through c-Jun-dependent Ubiquitin-proteasome Pathways in Regulating Ras Transformation and Stress Response. Journal of Biological Chemistry. 2007; 282: 31398–31408.
[83]
Alam MS, Gaida MM, Bergmann F, Lasitschka F, Giese T, Giese NA, et al. Selective inhibition of the p38 alternative activation pathway in infiltrating T cells inhibits pancreatic cancer progression. Nature Medicine. 2015; 21: 1337–1343.
[84]
Machado TR, Machado TR, Pascutti PG. The p38 MAPK Inhibitors and their Role in Inflammatory Diseases. ChemistrySelect. 2021; 6: 5729–5742.
[85]
Schreiber S, Feagan B, D’Haens Gea. Oral p38 mitogen-activated protein kinase inhibition with BIRB 796 for active Crohn’ disease: a randomized, double-blind, placebo-controlled trial. Clinical Gastroenterology Hapatology. 2006; 4: 325–334.
Share
Back to top