Next Article in Journal
Green Tea in Reproductive Cancers: Could Treatment Be as Simple?
Previous Article in Journal
Evaluation of Exposure Doses of Elective Nodal Irradiation in Chemoradiotherapy for Advanced Esophageal Cancer
Previous Article in Special Issue
Radiotherapeutic Strategies to Overcome Resistance of Breast Cancer Brain Metastases by Considering Immunogenic Aspects of Cancer Stem Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

P38 MAPK and Radiotherapy: Foes or Friends?

by
Natalia García-Flores
1,†,
Jaime Jiménez-Suárez
1,†,
Cristina Garnés-García
1,†,
Diego M. Fernández-Aroca
1,†,
Sebastia Sabater
1,2,
Ignacio Andrés
1,2,
Antonio Fernández-Aramburo
1,3,
María José Ruiz-Hidalgo
1,4,
Borja Belandia
5,
Ricardo Sanchez-Prieto
1,5,6,* and
Francisco J. Cimas
1,4,*
1
Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
2
Servicio de Oncología Radioterápica, Complejo Hospitalario Universitario de Albacete, 02006 Albacete, Spain
3
Servicio de Oncología Médica, Complejo Hospitalario Universitario de Albacete, 02006 Albacete, Spain
4
Departamento de Química Inorgánica, Orgánica y Bioquímica, Área de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
5
Departamento de Biología del Cáncer, Instituto de Investigaciones Biomédicas ‘Alberto Sols’ (CSIC-UAM), Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, 28029 Madrid, Spain
6
Departamento de Ciencias Médicas, Facultad de Medicina, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2023, 15(3), 861; https://doi.org/10.3390/cancers15030861
Submission received: 3 December 2022 / Revised: 16 January 2023 / Accepted: 24 January 2023 / Published: 30 January 2023
(This article belongs to the Special Issue Combating Tumor Radioresistance)

Abstract

:

Simple Summary

p38 MAPKs comprise a family of key proteins that regulate the stress response of human cells, controlling relevant biological phenomena such as the cell cycle. As a result, their role in the cellular response to ionizing radiation and radiotherapy is crucial. In this review, we provide in-depth insights into what is known about the whole protein family’s role in response to radiation and the implications for radiotherapy.

Abstract

Over the last 30 years, the study of the cellular response to ionizing radiation (IR) has increased exponentially. Among the various signaling pathways affected by IR, p38 MAPK has been shown to be activated both in vitro and in vivo, with involvement in key processes triggered by IR-mediated genotoxic insult, such as the cell cycle, apoptosis or senescence. However, we do not yet have a definitive clue about the role of p38 MAPK in terms of radioresistance/sensitivity and its potential use to improve current radiotherapy. In this review, we summarize the current knowledge on this family of MAPKs in response to IR as well as in different aspects related to radiotherapy, such as their role in the control of REDOX, fibrosis, and in the radiosensitizing effect of several compounds.

1. Knowing the p38 MAPK Protein Family

Mitogen-activated MAPKs are a superfamily of evolutionarily conserved serine/threonine proline target kinases (-X-Ser/Thr-Pro-X-) whose members regulate signalling cascades that convert different extracellular stimuli into a wide range of biological responses, such as survival, proliferation, differentiation or apoptosis. Among the different stimuli to which they respond are growth factors, neurotransmitters, cytokines, hormones and cellular stress [1].
In mammals, more than 14 MAPKs have been characterised and they can be classified into two groups: conventional and atypical MAPKs. While conventional MAPKs form a three-level transduction cascade and all members possess the conserved threonine-X-tyrosine (Thr-X-Tyr) motif, this hierarchical organization is lost in the atypical MAPK subfamily. The conventional MAPK group consists of extracellular signal-regulated protein kinase 1/2 (ERK 1/2), c-Jun amino (N)-terminal kinases 1–3 (JNK 1/2/3), p38 MAPKs (isoforms α, β, γ, δ), and the more recently discovered extracellular signal-regulated protein kinase 5 (ERK5). Atypical MAPKs include ERK 3/4, ERK7, ERK8 and Nemo-like kinase (NLK), whose activation cascade remains to be elucidated [2]. Regarding conventional MAPKs, the first discovered were the extracellular signal-regulated kinases (ERK) 1 and 2, which are activated mainly by growth factors, such as epidermal growth factor (EGF) or nerve growth factor (NGF) [3]. Jun N-Terminal Kinase (JNK) modules are mostly activated under conditions of cellular stress, including oxidative stress, cytokines, and UV radiation [4]. The p38 MAPKs are activated in response to different stimuli, such as oxidative stress, hypoxia, and osmotic and thermal shock, among others [5]. Finally, the most recently described module is ERK5, also known as Big MAPK1 (BMK1), whose activity increases in response to growth factors, serum, oxidative stress, and hyperosmolarity (for a review see [6]).

2. The p38 MAPK Pathway

The MAPK family is composed of four distinct proteins—widely labelled as isoforms—encoded by four genes: p38α (38 kDa, MAPK14 gene, 6p21.31), p38β (41.4 kDa, MAPK11 gene, 22q.13.33), p38ϒ (41.9 kDa, MAPK12 gene, 22q.13.33), and p38δ (42.1 kDa, MAPK13 gene, 6p21.31), and we refer to the four p38s collectively as p38 MAPK from now on (Figure 1). All four isoforms contain a conserved Thr-Gly-Tyr (TGY) motif that allows activation by dual phosphorylation of Thr and Tyr residues [7]. This family can be divided into two subgroups according to the percentage of homology in their amino acid sequence: p38α and p38β isoforms have 75% homology, while p38ϒ and p38δ share around 62% homology to p38α [8]. Furthermore, pharmacologically, they can be divided according to their sensitivity to pyridinyl imidazoles; p38α and p38β present a marked sensitivity to SB203580, while p38ϒ and p38δ are not affected by this compound [9]. In addition, each of the p38 MAPKs has a differential expression pattern in tissues, as well as a different substrate specificity, which may explain the diverse implications of each member in different biological processes as in the case of hematopoiesis [10]. p38α, the most abundant, and p38β are ubiquitously expressed while p38ϒ and p38δ show tissue-specific expression. p38ϒ is mainly expressed in skeletal muscle [11] and p38δ is expressed in the pancreas, kidney, small intestine, testis and lung [12]. As for its subcellular localization, p38 is ubiquitous, found both in the nucleus and in the cytoplasm under physiological conditions [13]. Following activation, accumulation of the p38α isoform has been observed in the nucleus, for example, after DNA damage [14,15]. It is also intriguing how the subcellular localization of p38α changes in response to different stimuli, as is its dependency on its own substrates, as in the case of MAPKAPK2, which is able to drive active p38α from the nucleus to the cytoplasm, thus explaining how it can phosphorylate various substrates in both compartments [14].
The best characterized member is p38α, as it was first identified in 1994 by observing tyrosine phosphorylation following endotoxin treatment, osmotic stress [16] and heat shock [17]. p38α has been identified as a key protein for the maintenance of tissue homeostasis, with involvement in different processes, ranging from differentiation to inflammatory response, and whose deregulation is implicated in several pathologies, such as neurodegenerative disorders and cancer (for a review see [18]). Despite its high homology, p38β cannot perform p38α-specific functions in embryonic development [19], and its role in acute or chronic inflammatory diseases seems to be minor [20]. Minority isoforms are less studied, although it has been recently demonstrated that their apparent secondary role is not so, with involvement in a wide range of pathologies such as cancer [21] and fungal infections [22], and whose relevance will undoubtedly increase in the coming years [23].
The p38 MAPK pathway consists of a three-kinase module in which a MAPKKK activates a MAPKK that in turn activates the MAPK by dual phosphorylation in the Thr-X-Tyr domain. There are different MAPKKKs capable of activating p38-dependent signaling, such as ASK1, TAK1, TAO and MLK3, although, in general, these MAPKKKs are promiscuous and capable of activating the JNK pathway as well [24]. In either case, canonical activation of p38 MAPK occurs through dual phosphorylation of the Thr and Tyr residues in the TGY motif by MKK3 and MKK6 kinases, leading to a conformational change that enhances protein activity and substrate recognition [25]. Although both MKK3 and MKK6 can potentially activate any of the four p38 kinases, there is evidence of some specificity between the different p38 isoforms and these MAPKKs [26] (Figure 1). However, other mechanisms of p38α activation by alternative phosphorylations have been described, such as phosphorylation at Tyr 323 by Zap70 kinase in Th1 lymphocytes following TCR receptor stimulation, allowing p38 to autophosphorylate and become self-activated [27], or activation through the ATR pathway, which is activated in response to DNA damage in cells where Cdc7 is depleted [28]. Once activated, its phosphorylation capacity will depend on its specific substrate binding sites (CD domains), characterized by the presence of hydrophobic and negatively charged residues, the subcellular location and the substrate concentration [29,30]. In fact, the sheer number of substrates may explain the plethora of effects in which p38 MAPK is involved (for a review see [31]).
Signal termination is due to a process of dephosphorylation. The involvement of the MAPK dual specific phosphatases (DUSP) has been described [32], with DUSP-1 probably being the best example [33]. Although it is not clear whether there is a specific phosphatase for each MAPK, a higher affinity for p38 MAPK has been described for some phosphatases, as in the case of PPMD1 [34], PP2A [35], and more recently DUSP-16, that is shared with JNK and seems to be implicated in cancer [36].

3. p38 MAPK and Cancer

The p38 MAPK pathway plays an important role in cancer initiation, progression and metastasis and has become a putative target for cancer therapy [37]. p38α is able to inhibit tumor proliferation, behaving as a tumor suppressor [38,39] through its ability to induce cell cycle arrest and senescence in response to stress stimuli [40]. In fact, it has been shown that its chemical inhibition can block tumor growth in vivo [41]. However, recent evidence demonstrates a dual role for p38α, which may behave as a tumor suppressor or as an oncogenic factor depending on the stage at which inactivation occurs [42] and with important implications for the metastatic process [43].
The role of p38α in cell cycle regulation is critical to understanding its involvement in cancer. It has been described how p38α can induce G1/S checkpoint arrest by downregulation of cyclin D1 through activation of HBP1 [44], or via direct phosphorylation of pRB, thereby increasing its affinity for the transcription factor E2F [45]. Furthermore, in contexts of DNA damage, it is known that p38α can be crucial in the activation of p53, controlling the expression of p21 and thus stopping the cell cycle in G1/S [46]. Nonetheless, p38α can also mediate G2/M arrest through targets as CDC25 or MK2 in response to DNA damage triggered by UV radiation or doxorubicin [47,48].
Its role in the tumor microenvironment is also relevant, as p38α signaling promotes tumorigenesis in lung cancer by generating a hyaluronan niche, leading to early activation of stromal fibroblasts and proliferation of cancer cells [49]. In breast cancer-associated fibroblasts, p38α promotes the activation of epithelial–mesenchymal transition (EMT) factors, such as fibronectin, N-cadherin and vimentin [50]. In addition, p38α has been proposed as a key factor in the control of other cell lineages involved in the temporal microenvironment, such as macrophages [51,52] or T cells [53].
Our knowledge of the role of the other p38 isoforms in cancer is much more limited and it is only now that we are beginning to identify their possible implications during tumorigenesis [54,55], with implications in processes such as inflammation [56] or immune system response [57]. In fact, they are already known to play a critical role in different types of tumors such as liver [21] and pancreatic cancer [58]. Those isoforms are even emerging as potential diagnostic markers in different types of tumors such as colon cancer [59], esophageal squamous cell carcinoma [60] or ovarian cancer [61].

4. p38 and UV Radiation

Ultraviolet (UV) radiation has been the most relevant type of radiation to which living beings have been naturally exposed and in which p38α plays a fundamental role [62]. Early studies showed that p38 MAPK was a UV-sensitive kinase [63]. p38α regulates cell cycle checkpoints in response to ultraviolet radiation by activating some of its substrates, such as CDC25 or MK2 [47,64], or more recently via its control over Claspin [65]. In addition, p38α is also involved in the repair of DNA damage caused by UV radiation. Thus, rapid activation of the p38α pathway also modifies the ubiquitination of substrates such as DDB2, facilitating the recruitment of XPC, which mediates DNA damage repair [66], or in the case of NR4A2, whose localization to sites of DNA damage is dependent on p38α signaling [67]. Although it is not known whether the activation of p38α is due to direct DNA damage or is a result of other effects generated by UV radiation, it seems to be clear that it is a determinant of cell fate [68]. In fact, in response to UV radiation, the activation of survival pathways such as AKT2 results in the attenuation of JNK and p38 activation [69], causing a decrease in UV-induced cell death and confirming the essential role of p38α in this process.

5. The P38 MAPK Pathway and Ionizing Radiation

Exposure to IR causes a simultaneous activation of ERK1/2, p38 MAPK, JNK [70] and also ERK5, which has recently been described as being activated in response to radiation [71,72]. This activation can be of varying intensity over time and depending on the cell type [73]. The ERK and JNK pathways appear to be in dynamic equilibrium following exposure to ionizing radiation, with the ERK pathway acting in a pro-survival role, inhibiting the JNK pathway, which acts in a pro-apoptotic role [74]. The activation of p38α by ionizing radiation was already described in the mid-1990s [75] and has been corroborated in vivo in different models [76,77]. This activation is mediated by some of its MAPKKKs, such as TAO or ASK1 [78,79]. However, it has not been specifically described which of its MAPKKs (MKK3/6) are essential for p38 MAPK activation, although it is foreseeable that both of them could be activated by IR, as occurs with most of the stimuli [26].

5.1. p38 MAPK and Cell Cycle in Response to IR

Activation of p38α in response to radiation results in a cellular response, leading to its translocation to the nucleus [15] and a regulation of cell cycle progression. In this sense, p38α mediates the blockade at different checkpoints such as G2/M via CDC25 [80] in which p38α activation could be mediated by TAO kinases (78). This response promotes cell cycle arrest, which, if it becomes permanent, ultimately triggers cell death [81]. p38 MAPK is also involved in the regulation of the G1/S checkpoint in response to IR through the phosphorylation of HuR, which stabilizes and triggers the cytoplasmic accumulation of p21/WAF in a p53-independent manner (Figure 2), thus mediating cell cycle arrest at this checkpoint [82]. However, it is not only p38α that has been implicated in cell cycle control in response to radiation; the MKK6-p38ϒ axis may also be involved in G2 phase control in an ATM-dependent manner [83]. It is striking that the two remaining p38 isoforms, p38β and p38δ, have not been implicated in cell cycle control in response to IR, which could indicate a certain specificity or different roles for the diverse members of the family in response to IR.

5.2. p38 MAPK, ROS and Cell Fate after IR Exposure

Another effect of IR is the generation of reactive oxygen species (ROS). The role of p38α in oxidative metabolism and ROS generation has been extensively studied both within and outside the context of radiotherapy [84,85]. In fact, it is known that inhibition of p38α can affect key molecules in redox balance. It has been described how p38 MAPK is a critical mediator in radioresistance associated with COX-2 in breast cancer [86]. Furthermore, recent evidence linked P38 MAPK with DRP1, a key effector of COX2 signaling in response to IR, mediated by mitochondrial fragmentation and TFAM upregulation [86,87]. The effect of p38 MAPK on ROS signaling could explain the radiosensitizing effect of compounds like Gliotoxin [88].
In addition, the p38 MAPK pathway is a critical player in cell fate after IR exposure. In this regard, p38α-dependent signaling mediates the induction of apoptosis in response to IR in different experimental models [77,89,90,91,92] via its control of key proteins in the apoptotic process [93]. Moreover, p38 is the target of proteins that block the induction of apoptosis in response to IR, such as Mast1 or COX-2 [86,94], and it has even been proposed to also be a target of miRNAs implicated in the induction of apoptosis and senescence associated with IR [95]. However, other studies demonstrate a cytoprotective role for p38 MAPK in response to IR [95,96], or even a lack of involvement [97], suggesting that the role of p38 MAPK may be different depending on the model under study. In any case, the vast majority of studies indicate a pro-apoptotic role for p38 MAPK-mediated signaling, especially in situations of genotoxic stress [98]. Another mechanism of cellular response to IR is the induction of senescence [99]. In this context, the inhibition of the ERK and p38 MAPK pathways leads to the blocking of IR-induced β-galactosidase activity by reducing the generation of ROS [74], with p16 and p53 being two of the main effectors of p38α in the regulation of senescent phenotypes [100]. It has even been proposed that some miRNAs that mediate the induction of senescence after IR exposure do so via p38α, as in the case of miR-155 [101]. In addition, a recent study points to p38β as a mediator of the cellular response to IR through modulation of the radiation-associated senescence process [102]. In summary, there seems to be a clear consensus that the lack of signaling via p38 MAPK promotes radiation-associated senescence. Finally, in terms of cell fate, autophagy is another critical response to IR [103]. The role of p38 MAPK has not yet been characterized in autophagy triggered by IR, although in response to other genotoxic agents, such as 5-FU [104], selenite [105] or irinotecan [106], this signaling pathway has been clearly implicated. It is therefore possible that a link between p38 MAPK and radiation-triggered autophagy could be established in the future.

5.3. P38 MAPK and Epithelial–Mesenchymal Transition in Response to IR

The effects of p38 MAPK are not just limited to control of cell death mechanisms triggered by IR. Indeed, it has been implicated in the epithelial–mesenchymal transition (EMT) associated with IR, a key process in radioresistance [107]. In this regard, it was previously shown how p38 MAPK controls migration, a biological process associated with EMT, in response to IR in the A549 cell line [108]. It was later demonstrated how, in hepatocarcinoma cell lines, a pattern of reduced expression of E-cadherin and enhanced expressions of N-cadherin, Vimentin and Snail was found after IR exposure, which is consistent with EMT induction [109]. This effect seems to be mediated by the induction of a novel hydrogen sulfide signaling pathway that upregulates the expressions of H2S-producing proteins such as cysthionine-γ-lyase and cystathionine-β-synthase, which mediate p38α activation in response to IR. In fact, the authors showed how cysthionine-γ-lyase genetic ablation was able to prevent p38α activation and EMT in response to IR, and, more specifically, chemical inhibition of p38 MAPK was able to block IR-induced EMT in these hepatocarcinoma cell lines. In this regard, it has been proposed that MK2, one of the direct substrates of p38 MAPK, is a critical mediator in the control of IR-associated EMT exerted by p38 MAPK in head and neck cancer models [110]. Of note, it has been observed that both modulation of p38α activation and IR-induced EMT could be controlled by PARP1, as was indicated by the use of olaparib [111,112]. Therefore, all this evidence suggests that p38 MAPK is a key regulator of EMT associated with IR treatment, which renders unwanted associated effects such as the induction of metastasis or fibrosis.

5.4. Regulation of p38 MAPK Signaling in Response to IR

Nonetheless, it should be noted that not only the activation of p38 MAPK, but also its deactivation, is important in the response to IR. In fact, phosphatases that dephosphorylate the p38 MAPKs also appear to play an important role in the response to radiotherapy. An example of this is DUSP-1, as high expression of this phosphatase is correlated with a decrease in the effectiveness of radiotherapy by promoting DNA repair after exposure to IR through dephosphorylation of H3S10 [113]. In addition, silencing of this phosphatase leads to decreased colony formation and improved radiosensitivity in breast cancer stem cells [114]. In the case of phosphatase DUSP-16, its high expression appears to markedly decrease radiotherapy-induced apoptosis [115]. On the other hand, PP2A phosphatase is downregulated in many tumor types [116], although it has been reported that after inhibition of this phosphatase many tumors show slower growth as well as increased cell death by apoptosis and increased radiosensitivity [117]. The pharmacological approach, therefore, seems promising for regulating the phosphorylation of this pathway and thus modulating the radioresistance of different tumor types.
However, it is important to note that activation of p38 MAPK by IR is not universal. Thus, while the studies mentioned above indicate a clear activation of the pathway, others show very weak or no activation after exposure to ionizing radiation, as for example in the presence of viral genes [118]. In fact, the effect of pathway regulation could be different depending on the genetic context or the expression of certain molecules. For example, in the case of the RAS oncogenes, the expression of Ki-Ras causes a clear activation of p38 and radiosensitivity, whereas with expression of Ha-Ras, no activation of p38 is observed [119]. Another example could be TNF-α-associated radiosensitivity in lung cancer, which appears to be mediated by activation of stress response pathways, such as p38α [120]. Notably, in a murine screen for the main proteins involved in the radiation response, the p38α protein was predicted to be one of the key proteins in this process, despite the lack of changes in its expression level [121]. However, other proteomics-based screenings have shown that JNK also plays a fundamental role in this response [97]. We can therefore conclude that p38 MAPK-mediated signaling is an important effector of the cellular response to IR, but this role would depend on the cellular and genetic context in which it occurs.

6. Role of p38 MAPK in the Irradiation of Healthy Tissue

Not only does p38 MAPK mediate the effects on tumor tissue but also part of the effects of radiation on adjacent tissues or even further away from the tumor, which is known as the abscopal effect [122] and has been related to p38 MAPK. The exposure to low doses of radiation, such as at the tumor periphery in radiotherapy treatments, can lead to increased cell migration and invasion mediated by p38 MAPK and regulated by connexin 43 expression, which may promote metastasis formation [123]. In fact, in cells that have not been directly subjected to radiation (“bystander” cells), the activity of p38α and its substrates is increased [124]. In this regard, a recent study shows that irradiation of the thorax of mice causes a decrease in their reproductive capacity through activation of the TNFα/p38α axis, constituting another example of an abscopal effect mediated by this protein [125]. Indeed, p38α appears to play an important role in certain cellular processes related to the response to IR and not directly linked to tumor control. In endothelial cells, which are key in radiotherapy treatment for both tumor control and side effects, IR induces death by apoptosis mainly through the activation of p38α [92]. Radiation-induced cardiac damage is one of the causes of death in cancer patients receiving radiotherapy. Treatment with the cardio-protectant L-carnitine was shown to reduce radiation-induced alterations in cardiac function by decreasing myocyte apoptosis and ROS production, effects that disappeared when treated with the p38α/β inhibitor SB203580 [126]. In addition, treatment with SB203580 appears to block IL-8 overexpression after radiation exposure, clarifying the possible mechanism of action of radiation pneumonitis [127]. Furthermore, the role of p38 in myelosuppression with radiotherapy has been studied using p38 MAPK inhibitors. For example, SB203580 attenuates radiation-induced damage and senescence in hematopoietic stem cells [128] and inhibits the ROS–p38 MAPK pathway, although this is not sufficient to ameliorate long-term myelosuppression induced by total body irradiation [129]. Another study showed that adjuvant treatment with SB2003580 together with granulocyte colony-stimulating factor was able to increase hematopoiesis, mitigating the effects of total body irradiation in mice [130]. Fibrosis associated with radiation exposure in healthy tissues, as in the case of the lung [131], is one of the unresolved issues in radiotherapy. In this sense, it has been described how p38 MAPK may play an important role and its inhibition may be decisive in radiation-associated fibrosis, as has been proved in other pathological contexts such as renal fibrosis [132]. Therefore, the role of p38 MAPK in this unwanted effect of radiation should be studied in depth. In fact, there is already evidence to support the role of p38α inhibition in the control of radiation-associated fibrosis, as in the case of amifostine analogues [133]. Another type of secondary effect associated with radiotherapy is intestinal damage [134,135]. Thus, DUSP-16, a phosphatase specific to JNK and p38α [136], is implicated in this adverse effect of radiotherapy, as indicated by the use of compounds such as flagellin or rheinic acid [115,137]. Even differential activation of MAPKs, including p38 MAPK, could explain the different intestinal damage observed between sexes in experimental models [138]. In summary, p38 MAPK may play a decisive role in several aspects of radiotherapy treatment in healthy tissue, which in many cases becomes a limiting step that can lead to treatment failure.

7. p38 MAPK and Radiosensitivity

As mentioned above, the data available to date do not allow a definitive role to be assigned to the p38 MAPK pathway in terms of radioresistance or radiosensitivity. There are many studies in which inhibition/lack of activation correlates with a radioresistant phenotype and others in which it has no effect or even the opposite. What there does seem to be more consensus on is the potential of p38 MAPK-dependent signaling to mediate the radiosensitizing effect of different drugs. For example, p38α is fundamental to the radiosensitizing effects of 5-FU [139,140]. These observations may have certain clinical implications, as demonstrated by a retrospective analysis of 74 patients with rectal cancer that revealed a strong correlation between p38 MAPK levels and improved prognosis and response to treatment with radiotherapy combined with chemotherapy, such as 5-FU or FOLFOX, which led to increased cell death [141]. Another clear example is that of gemcitabine, although in this case it is not p38α but p38β which seems to mediate this radiosensitizing effect [142]. In addition, the p38 MAPK pathway also mediates the effect of radiosensitizers of a very different nature. One study showed that the combined treatment of arsenic trioxide with IR caused an increase in apoptosis through the generation of ROS and the activation of p38α, leading to a synergistic effect that enhanced the response of leukemia cells to IR [143]. Kale et al. have shown that the combined treatment of PU-H71, a novel Hsp90 chaperone inhibitor, with radiotherapy has a potent radiosensitizing effect that correlates with increased p38α activation [144]. In addition, the combination of berberine with radiation appears to have a synergistic effect via p38α and ROS generation [145]. Moreover, a recent study presented a synthetic alkylating agent, LQB-118, as a radiosensitizing agent in glioblastoma, which operated by reducing the total expression and phosphorylation levels of p38 [146]. Similarly, luteolin, a flavonoid, appears to increase cell death by apoptosis in combination with IR through phosphorylation and subsequent activation of the p38 MAPK pathway, among other processes [147]. Treatment with biological therapies, such as unmethylated CpG dinucleotides, has been shown to cause activation of TLR9 as well as activation of p38α, leading to increased G2/M arrest and cell death by apoptosis [148]. Another study showed how treatment of breast cancer cells with a phospholipase D inhibitor increased phosphorylation of the p38 MAPK pathway in response to radiation, and inhibition of its activation with SB203580 resulted in the loss of the increased cell death caused by the phospholipase D inhibitor in combination with radiotherapy [149]. Finally, p38α is responsible for the radiosensitivity/resistance associated with other genes; recent examples include the case of Anexin2 [81], NA methyltransferase 3A and B [150], the ubiquitin protein ligase E3 component n-recognin (UBR5) [151], or more recently, the Anterior Gradient Protein 2 homolog (AGR-2) [152]. Furthermore, p38α has been proposed to mediate the radiosensitivity associated with lncRNAs, such as TPTEP1 [153], or miRNAs, such as miRNA-153 [154]. Therefore, p38 MAPK signaling can be also a critical mediator of the radiosensitizing effects associated with different compounds, genes and other regulatory molecules, reinforcing its importance in current radiotherapy.

8. Conclusions and Future Perspectives

All evidence points to an important role for the p38 MAPK signaling pathway in the response to IR, with possible clinical implications in radiotherapy. Research questions involve the role of the different isoforms, the cellular and genetic context that defines the role of p38 MAPK in the response to IR, the involvement of p38 MAPK in the different mechanisms of cell death triggered by IR, or the role in EMT associated to IR, among others. It would also be very interesting to know how p38 MAPK can control fibrosis or how p38 MAPK mediates intestinal and vascular damage in response to radiotherapy. However, the probably most relevant question encompassing all of the above is how can we improve current radiotherapy by modulating p38 MAPK expression or activity. The fact that p38 MAPK may be affecting radioresistance in irradiated tumor cells as well as its possible involvement in the effects of radiation on healthy tissue should be carefully considered for future radiotherapy treatments. To date, only one clinical trial using the highly selective p38α and p38β inhibitor ralimetinib with radiotherapy has been reported, showing good tolerability, low toxicity and encouraging results [155]. In this sense, the development of new specific inhibitors [156] or new PROTAC-based specific degraders [157] opens a new window to fully exploit the potential of p38 MAPK in radiotherapy.

Author Contributions

N.G.-F., J.J.-S., C.G.-G. and D.M.F.-A. contributed to Section 1 to Section 7 and thoroughly proofread the manuscript for its submission. A.F.-A. contributed to Section 7 and provided proofreading comments. S.S. and I.A. collaborated in the composition of Section 5, Section 6 and Section 7 and provided proofreading comments. B.B. and M.J.R.-H. collaborated in Section 1, Section 2 and Section 3 and thoroughly proofread the manuscript for its submission. R.S.-P. and F.J.C. designed and coordinated the whole review, collaborated in composing all the sections, created figures, edited and thoroughly proofread the manuscript for its submission. All authors have read and agreed to the published version of the manuscript.

Funding

This work has been supported by grant PID2021-122222OB-I00 funded by MCIN/AEI /10.13039/501100011033/ and by FEDER A way to make Europe. F.J. Cimas is funded by contracts for post-doctoral researchers for scientific excellence in the development of the Plan Propio I+D+i, co-financed by the European Social Fund Plus (ESF+). N. García-Flores is funded by the “Investigo Programme” for hiring young job seekers to carry out research and innovation initiatives within the framework of the Recovery, Transformation and Resilience Plan, Next Generation EU, which is financed by the European Union, called by Order 190/2021 of the Regional Ministry of Economy, Business and Employment of the Regional Government of Castilla-La Mancha on 22 December. J. Jiménez-Suárez is funded by the “Contrato predoctoral para la formación de personal investigador” en el marco del plan propio de I+D+I UCLM 2020-PREDUCLM-15144. co-financed by the European Social Fund Plus (ESF+).

Acknowledgments

We want to acknowledge Fundación Leticia Castillejo Castillo and ACEPAIN for their support in our research. We would like to apologize for not citing the work of several laboratories due to the space limitations.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Cargnello, M.; Roux, P.P. Activation and function of the MAPKs and their substrates, the MAPK-activated protein kinases. Microbiol. Mol. Biol. Rev. MMBR 2011, 75, 50–83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Coulombe, P.; Meloche, S. Atypical mitogen-activated protein kinases: Structure, regulation and functions. Biochim. Biophys. Acta 2007, 1773, 1376–1387. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Boulton, T.G.; Yancopoulos, G.D.; Gregory, J.S.; Slaughter, C.; Moomaw, C.; Hsu, J.; Cobb, M.H. An Insulin-Stimulated Protein Kinase Similar to Yeast Kinases Involved in Cell Cycle Control. Science 1990, 249, 64–67. [Google Scholar] [CrossRef] [PubMed]
  4. Bogoyevitch, M.A.; Ngoei, K.R.; Zhao, T.T.; Yeap, Y.Y.; Ng, D.C. c-Jun N-terminal kinase (JNK) signaling: Recent advances and challenges. Biochim. Biophys. Acta (BBA)-Proteins Proteom. 2010, 1804, 463–475. [Google Scholar] [CrossRef] [PubMed]
  5. Sanz-Ezquerro, J.J.; Cuenda, A. p38 Signalling Pathway. Int. J. Mol. Sci. 2021, 22, 1003. [Google Scholar] [CrossRef]
  6. Paudel, R.; Fusi, L.; Schmidt, M. The MEK5/ERK5 Pathway in Health and Disease. Int. J. Mol. Sci. 2021, 22, 7594. [Google Scholar] [CrossRef]
  7. Cuenda, A.; Rousseau, S. p38 MAP-kinases pathway regulation, function and role in human diseases. Biochim. Biophys. Acta 2007, 1773, 1358–1375. [Google Scholar] [CrossRef] [Green Version]
  8. Risco, A.; Cuenda, A. New Insights into the p38γ and p38δ MAPK Pathways. J. Signal Transduct. 2011, 2012, 520289. [Google Scholar] [CrossRef] [Green Version]
  9. Cuenda, A.; Rouse, J.; Doza, Y.N.; Meier, R.; Cohen, P.; Gallagher, T.F.; Young, P.R.; Lee, J.C. SB 203580 is a specific inhibitor of a MAP kinase homologue which is stimulated by cellular stresses and interleukin-1. FEBS Lett. 1995, 364, 229–233. [Google Scholar]
  10. Uddin, S.; Ah-Kang, J.; Ulaszek, J.; Mahmud, D.; Wickrema, A. Differentiation stage-specific activation of p38 mitogen-activated protein kinase isoforms in primary human erythroid cells. Proc. Natl. Acad. Sci. USA 2004, 101, 147–152. [Google Scholar] [CrossRef] [Green Version]
  11. Pogozelski, A.R.; Geng, T.; Li, P.; Yin, X.; Lira, V.A.; Zhang, M.; Chi, J.-T.; Yan, Z. p38gamma mitogen-activated protein kinase is a key regulator in skeletal muscle metabolic adaptation in mice. PLoS ONE 2009, 4, e7934. [Google Scholar] [CrossRef] [PubMed]
  12. Goedert, M.; Cuenda, A.; Craxton, M.; Jakes, R.; Cohen, P. Activation of the novel stress-activated protein kinase SAPK4 by cytokines and cellular stresses is mediated by SKK3 (MKK6); comparison of its substrate specificity with that of other SAP kinases. EMBO J. 1997, 16, 3563–3571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Raingeaud, J.; Gupta, S.; Rogers, J.S.; Dickens, M.; Han, J.; Ulevitch, R.J.; Davis, R.J. Pro-inflammatory Cytokines and Environmental Stress Cause p38 Mitogen-activated Protein Kinase Activation by Dual Phosphorylation on Tyrosine and Threonine. J. Biol. Chem. 1995, 270, 7420–7426. [Google Scholar] [CrossRef] [Green Version]
  14. Ben-Levy, R.; Hooper, S.; Wilson, R.; Paterson, H.F.; Marshall, C.J. Nuclear export of the stress-activated protein kinase p38 mediated by its substrate MAPKAP kinase-2. Curr. Biol. 1998, 8, 1049–1057. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Wood, C.D.; Thornton, T.M.; Sabio, G.; Davis, R.A.; Rincon, M. Nuclear Localization of p38 MAPK in Response to DNA Damage. Int. J. Biol. Sci. 2009, 5, 428–437. [Google Scholar] [CrossRef] [Green Version]
  16. Han, J.; Lee, J.D.; Bibbs, L.; Ulevitch, R.J. A MAP Kinase Targeted by Endotoxin and Hyperosmolarity in Mammalian Cells. Science 1994, 265, 808–811. [Google Scholar] [CrossRef]
  17. Rouse, J.; Cohen, P.; Trigon, S.; Morange, M.; Alonso-Llamazares, A.; Zamanillo, D.; Hunt, T.; Nebreda, A.R. A novel kinase cascade triggered by stress and heat shock that stimulates MAPKAP kinase-2 and phosphorylation of the small heat shock proteins. Cell 1994, 78, 1027–1037. [Google Scholar] [CrossRef]
  18. Canovas, B.; Nebreda, Á.R. Diversity and versatility of p38 kinase signalling in health and disease. Nat. Rev. Mol. Cell Biol. 2021, 22, 346–366. [Google Scholar] [CrossRef]
  19. del Barco Barrantes, I.; Coya, J.M.; Maina, F.; Arthur, J.S.C.; Nebreda, Á.R. Genetic analysis of specific and redundant roles for p38α and p38β MAPKs during mouse development. Proc. Natl. Acad. Sci. USA 2011, 108, 12764–12769. [Google Scholar] [CrossRef] [Green Version]
  20. O’Keefe, S.J.; Mudgett, J.S.; Cupo, S.; Parsons, J.N.; Chartrain, N.A.; Fitzgerald, C.; Chen, S.-L.; Lowitz, K.; Rasa, C.; Visco, D.; et al. Chemical Genetics Define the Roles of p38α and p38β in Acute and Chronic Inflammation. J. Biol. Chem. 2007, 282, 34663–34671. [Google Scholar] [CrossRef] [Green Version]
  21. Tomás-Loba, A.; Manieri, E.; González-Terán, B.; Mora, A.; Leiva-Vega, L.; Santamans, A.M.; Romero-Becerra, R.; Rodríguez, E.; Pintor-Chocano, A.; Feixas, F.; et al. p38γ is essential for cell cycle progression and liver tumorigenesis. Nature 2019, 568, 557–560. [Google Scholar] [CrossRef] [PubMed]
  22. Alsina-Beauchamp, D.; Escós, A.; Fajardo, P.; González-Romero, D.; Díaz-Mora, E.; Risco, A.; Martín-Serrano, M.A.; Del Fresno, C.; Dominguez-Andrés, J.; Aparicio, N.; et al. Myeloid cell deficiency of p38γ/p38δ protects against candidiasis and regulates antifungal immunity. EMBO Mol. Med. 2018, 10, e8485. [Google Scholar] [CrossRef] [PubMed]
  23. Cuenda, A.; Sanz-Ezquerro, J.J. p38γ and p38δ: From Spectators to Key Physiological Players. Trends Biochem. Sci. 2017, 42, 431–442. [Google Scholar] [CrossRef] [PubMed]
  24. Ichijo, H.; Nishida, E.; Irie, K.; ten Dijke, P.; Saitoh, M.; Moriguchi, T.; Takagi, M.; Matsumoto, K.; Miyazono, K.; Gotoh, Y. Induction of apoptosis by ASK1, a mammalian MAPKKK that activates SAPK/JNK and p38 signaling pathways. Science 1997, 275, 90–94. [Google Scholar] [CrossRef]
  25. Dérijard, B.; Raingeaud, J.; Barrett, T.; Wu, I.-H.; Han, J.; Ulevitch, R.J.; Davis, R.J. Independent Human MAP-Kinase Signal Transduction Pathways Defined by MEK and MKK Isoforms. Science 1995, 267, 682–685. [Google Scholar] [CrossRef]
  26. Remy, G.; Risco, A.M.; Iñesta-Vaquera, F.A.; González-Terán, B.; Sabio, G.; Davis, R.J.; Cuenda, A. Differential activation of p38MAPK isoforms by MKK6 and MKK3. Cell. Signal. 2010, 22, 660–667. [Google Scholar] [CrossRef]
  27. Salvador, J.M.; Mittelstadt, P.R.; Guszczynski, T.; Copeland, T.D.; Yamaguchi, H.; Appella, E.; Fornace, A.J.; Ashwell, J.D. Alternative p38 activation pathway mediated by T cell receptor–proximal tyrosine kinases. Nat. Immunol. 2005, 6, 390–395. [Google Scholar] [CrossRef]
  28. Im, J.-S.; Lee, J.-K. ATR-dependent activation of p38 MAP kinase is responsible for apoptotic cell death in cells depleted of Cdc7. J. Biol. Chem. 2008, 283, 25171–25177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Tanoue, T.; Adachi, M.; Moriguchi, T.; Nishida, E. A conserved docking motif in MAP kinases common to substrates, activators and regulators. Nat. Cell Biol. 2000, 2, 110–116. [Google Scholar] [CrossRef]
  30. Tanoue, T.; Maeda, R.; Adachi, M.; Nishida, E. Identification of a docking groove on ERK and p38 MAP kinases that regulates the specificity of docking interactions. EMBO J. 2001, 20, 466–479. [Google Scholar] [CrossRef]
  31. Han, J.; Wu, J.; Silke, J. An overview of mammalian p38 mitogen-activated protein kinases, central regulators of cell stress and receptor signaling. F1000Research 2020, 9, 653. [Google Scholar] [CrossRef] [PubMed]
  32. Lang, R.; Raffi, F.A.M. Dual-Specificity Phosphatases in Immunity and Infection: An Update. Int. J. Mol. Sci. 2019, 20, 2710. [Google Scholar] [CrossRef] [Green Version]
  33. Shen, J.; Zhang, Y.; Yu, H.; Shen, B.; Liang, Y.; Jin, R.; Liu, X.; Shi, L.; Cai, X. Role of DUSP1/MKP1 in tumorigenesis, tumor progression and therapy. Cancer Med. 2016, 5, 2061–2068. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Nahta, R.; Castellino, R.C. Phosphatase magnesium-dependent 1 δ (PPM1D), serine/threonine protein phosphatase and novel pharmacological target in cancer. Biochem. Pharmacol. 2021, 184, 114362. [Google Scholar] [CrossRef] [PubMed]
  35. Wang, Y.; Xia, Y.; Kuang, D.; Duan, Y.; Wang, G. PP2A regulates SCF-induced cardiac stem cell migration through interaction with p38 MAPK. Life Sci. 2017, 191, 59–67. [Google Scholar] [CrossRef]
  36. Shillingford, S.; Zhang, L.; Surovtseva, Y.; Dorry, S.; Lolis, E.; Bennett, A.M. A novel site on dual-specificity phosphatase MKP7/DUSP16 is required for catalysis and MAPK binding. J. Biol. Chem. 2022, 298, 102617. [Google Scholar] [CrossRef]
  37. García-Hernández, L.; García-Ortega, M.B.; Ruiz-Alcalá, G.; Carrillo, E.; Marchal, J.A.; García, M.Á. The p38 MAPK Components and Modulators as Biomarkers and Molecular Targets in Cancer. Int. J. Mol. Sci. 2021, 23, 370. [Google Scholar] [CrossRef]
  38. Ventura, J.J.; Tenbaum, S.; Perdiguero, E.; Huth, M.; Guerra, C.; Barbacid, M.; Pasparakis, M.; Nebreda, Á.R. p38α MAP kinase is essential in lung stem and progenitor cell proliferation and differentiation. Nat. Genet. 2007, 39, 750–758. [Google Scholar] [CrossRef]
  39. Hui, L.; Bakiri, L.; Mairhorfer, A.; Schweifer, N.; Haslinger, C.; Kenner, L.; Komnenovic, V.; Scheuch, H.; Beug, H.; Wagner, E.F. p38α suppresses normal and cancer cell proliferation by antagonizing the JNK–c-Jun pathway. Nat. Genet. 2007, 39, 741–749. [Google Scholar] [CrossRef]
  40. Wang, W.; Chen, J.X.; Liao, R.; Deng, Q.; Zhou, J.J.; Huang, S.; Sun, P. Sequential Activation of the MEK-Extracellular Signal-Regulated Kinase and MKK3/6-p38 Mitogen-Activated Protein Kinase Pathways Mediates Oncogenic ras-Induced Premature Senescence. Mol. Cell. Biol. 2002, 22, 3389–3403. [Google Scholar] [CrossRef] [Green Version]
  41. Gupta, J.; Igea, A.; Papaioannou, M.; Lopez-Casas, P.P.; Llonch, E.; Hidalgo, M.; Gorgoulis, V.G.; Nebreda, Á.R. Pharmacological inhibition of p38 MAPK reduces tumor growth in patient-derived xenografts from colon tumors. Oncotarget 2015, 6, 8539–8551. [Google Scholar] [CrossRef] [PubMed]
  42. Gupta, J.; del Barco Barrantes, I.; Igea, A.; Sakellariou, S.; Pateras, I.S.; Gorgoulis, V.G.; Nebreda, Á.R. Dual function of p38α MAPK in colon cancer: Suppression of colitis-associated tumor initiation but requirement for cancer cell survival. Cancer Cell 2014, 25, 484–500. [Google Scholar] [CrossRef] [Green Version]
  43. Urosevic, J.; Garcia-Albéniz, X.; Planet, E.; Real, S.; Céspedes, M.V.; Guiu, M.; Fernandez, E.; Bellmunt, A.; Gawrzak, S.; Pavlovic, M.; et al. Colon cancer cells colonize the lung from established liver metastases through p38 MAPK signalling and PTHLH. Nat. Cell Biol. 2014, 16, 685–694. [Google Scholar] [CrossRef] [PubMed]
  44. Thoms, H.C.; Dunlop, M.G.; Stark, L.A. p38-mediated inactivation of cyclin D1/cyclin-dependent kinase 4 stimulates nucleolar translocation of RelA and apoptosis in colorectal cancer cells. Cancer Res. 2007, 67, 1660–1669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Gubern, A.; Joaquin, M.; Marquès, M.; Maseres, P.; Garcia-Garcia, J.; Amat, R.; González-Nuñez, D.; Oliva, B.; Real, F.X.; de Nadal, E.; et al. The N-Terminal Phosphorylation of RB by p38 Bypasses Its Inactivation by CDKs and Prevents Proliferation in Cancer Cells. Mol. Cell 2016, 64, 25–36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Sanchez-Prieto, R.; Rojas, J.M.; Taya, Y.; Gutkind, J.S. A role for the p38 mitogen-acitvated protein kinase pathway in the transcriptional activation of p53 on genotoxic stress by chemotherapeutic agents. Cancer Res. 2000, 60, 2464–2472. [Google Scholar]
  47. Bulavin, D.V.; Higashimoto, Y.; Popoff, I.J.; Gaarde, W.A.; Basrur, V.; Potapova, O.; Appella, E.; Fornace, A.J. Initiation of a G2/M checkpoint after ultraviolet radiation requires p38 kinase. Nature 2001, 411, 102–107. [Google Scholar] [CrossRef]
  48. Reinhardt, H.C.; Aslanian, A.S.; Lees, J.A.; Yaffe, M.B. p53-Deficient Cells Rely on ATM- and ATR-Mediated Checkpoint Signaling through the p38MAPK/MK2 Pathway for Survival after DNA Damage. CCELL 2007, 11, 175–189. [Google Scholar] [CrossRef] [Green Version]
  49. Brichkina, A.; Bertero, T.; Loh, H.M.; Nguyen, N.T.M.; Emelyanov, A.; Rigade, S.; Ilie, M.; Hofman, P.; Gaggioli, C.; Bulavin, D.V. p38MAPK builds a hyaluronan cancer niche to drive lung tumorigenesis. Genes Dev. 2016, 30, 2623–2636. [Google Scholar] [CrossRef] [Green Version]
  50. Wen, S.; Hou, Y.; Fu, L.; Xi, L.; Yang, D.; Zhao, M.; Qin, Y.; Sun, K.; Teng, Y.; Liu, M. Cancer-associated fibroblast (CAF)-derived IL32 promotes breast cancer cell invasion and metastasis via integrin β3–p38 MAPK signalling. Cancer Lett. 2019, 442, 320–332. [Google Scholar] [CrossRef]
  51. Li, Z.; Liu, F.-Y.; Kirkwood, K.L. The p38/MKP-1 signaling axis in oral cancer: Impact of tumor-associated macrophages. Oral Oncol. 2020, 103, 104591. [Google Scholar] [CrossRef] [PubMed]
  52. Neamatallah, T. Mitogen-Activated Protein Kinase Pathway: A Critical Regulator in Tumor-associated Macrophage Polarization. J. Microsc. Ultrastruct. 2019, 7, 53–56. [Google Scholar] [CrossRef] [PubMed]
  53. Romagnani, A.; Rottoli, E.; Mazza, E.M.C.; Rezzonico-Jost, T.; De Ponte Conti, B.; Proietti, M.; Perotti, M.; Civanelli, E.; Perruzza, L.; Catapano, A.L.; et al. P2X7 Receptor Activity Limits Accumulation of T Cells within Tumors. Cancer Res. 2020, 80, 3906–3919. [Google Scholar] [CrossRef] [PubMed]
  54. Xu, W.; Liu, R.; Dai, Y.; Hong, S.; Dong, H.; Wang, H. The Role of p38γ in Cancer: From review to outlook. Int. J. Biol. Sci. 2021, 17, 4036–4046. [Google Scholar] [CrossRef]
  55. Roche, O.; Fernández-Aroca, D.M.; Arconada-Luque, E.; García-Flores, N.; Mellor, L.F.; Ruiz-Hidalgo, M.J.; Sánchez-Prieto, R. p38β and Cancer: The Beginning of the Road. Int. J. Biol. Sci. 2020, 21, 7524. [Google Scholar] [CrossRef] [PubMed]
  56. Qin, J.-Z.; Xin, H.; Qi, X.-M.; Chen, G. Isoform-specific and cell/tissue-dependent effects of p38 MAPKs in regulating inflammation and inflammation-associated oncogenesis. Front. Biosci. (Landmark Ed.) 2022, 27, 31. [Google Scholar] [CrossRef] [PubMed]
  57. Barrio, L.; Román-García, S.; Díaz-Mora, E.; Risco, A.; Jiménez-Saiz, R.; Carrasco, Y.R.; Cuenda, A. B Cell Development and T-Dependent Antibody Response Are Regulated by p38γ and p38δ. Front. Cell Dev. Biol. 2020, 8, 189. [Google Scholar] [CrossRef]
  58. Wang, F.; Qi, X.-M.; Wertz, R.; Mortensen, M.; Hagen, C.; Evans, J.; Sheinin, Y.; James, M.; Liu, P.; Tsai, S.; et al. p38γ MAPK Is Essential for Aerobic Glycolysis and Pancreatic Tumorigenesis. Cancer Res. 2020, 80, 3251–3264. [Google Scholar] [CrossRef]
  59. Fajardo, P.; Taskova, M.; Martín-Serrano, M.A.; Hansen, J.; Slott, S.; Jakobsen, A.K.; Wibom, M.-L.; Salegi, B.; Muñoz, A.; Barbachano, A.; et al. p38γ and p38δ as biomarkers in the interplay of colon cancer and inflammatory bowel diseases. Cancer Commun. 2022, 42, 897–901. [Google Scholar] [CrossRef]
  60. Zheng, S.; Yang, C.; Liu, T.; Liu, Q.; Dai, F.; Sheyhidin, I.; Lu, X. Clinicopathological significance of p38β, p38γ, and p38δ and its biological roles in esophageal squamous cell carcinoma. Tumour Biol. 2016, 37, 7255–7266. [Google Scholar] [CrossRef]
  61. Katopodis, P.; Kerslake, R.; Zikopoulos, A.; Beri, N.; Anikin, V. p38β-MAPK11 and its role in female cancers. J. Ovarian Res. 2021, 14, 84. [Google Scholar] [CrossRef]
  62. López-Camarillo, C.; Ocampo, E.A.; Casamichana, M.L.; Pérez-Plasencia, C.; Alvarez-Sánchez, E.; Marchat, L.A. Protein kinases and transcription factors activation in response to UV-radiation of skin: Implications for carcinogenesis. Int. J. Biol. Sci. 2012, 13, 142–172. [Google Scholar] [CrossRef] [PubMed]
  63. Liu, Y.; Guyton, K.Z.; Gorospe, M.; Xu, Q.; Lee, J.C.; Holbrook, N.J. Differential activation of ERK, JNK/SAPK and P38/CSBP/RK map kinase family members during the cellular response to arsenite. Free. Radic. Biol. Med. 1996, 21, 771–781. [Google Scholar] [CrossRef] [PubMed]
  64. Manke, I.A.; Nguyen, A.; Lim, D.; Stewart, M.Q.; Elia, A.E.H.; Yaffe, M.B. MAPKAP kinase-2 is a cell cycle checkpoint kinase that regulates the G2/M transition and S phase progression in response to UV irradiation. Mol. Cell 2005, 17, 37–48. [Google Scholar] [CrossRef]
  65. Ulsamer, A.; Martínez-Limón, A.; Bader, S.; Rodríguez-Acebes, S.; Freire, R.; Méndez, J.; de Nadal, E.; Posas, F. Regulation of Claspin by the p38 stress-activated protein kinase protects cells from DNA damage. Cell Rep. 2022, 40, 111375. [Google Scholar] [CrossRef] [PubMed]
  66. Zhao, Q.; Barakat, B.M.; Qin, S.; Ray, A.; El-Mahdy, M.A.; Wani, G.; Arafa, E.-S.; Mir, S.N.; Wang, Q.-E.; Wani, A.A. The p38 mitogen-activated protein kinase augments nucleotide excision repair by mediating DDB2 degradation and chromatin relaxation. J. Biol. Chem. 2008, 283, 32553–32561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Jagirdar, K.; Yin, K.; Harrison, M.; Lim, W.; Muscat, G.E.O.; Sturm, R.A.; Smith, A.G. The NR4A2 nuclear receptor is recruited to novel nuclear foci in response to UV irradiation and participates in nucleotide excision repair. PLoS ONE 2013, 8, e78075. [Google Scholar] [CrossRef] [Green Version]
  68. Bulavin, D.V.; Saito, S.; Hollander, M.C.; Sakaguchi, K.; Anderson, C.W.; Appella, E.; Fornace, A.J. Phosphorylation of human p53 by p38 kinase coordinates N-terminal phosphorylation and apoptosis in response to UV radiation. EMBO J. 1999, 18, 6845–6854. [Google Scholar] [CrossRef]
  69. Kim, M.-A.; Kim, H.-J.; Jee, H.J.; Kim, A.J.; Bae, Y.-S.; Bae, S.S.; Yun, J. Akt2, but not Akt1, is required for cell survival by inhibiting activation of JNK and p38 after UV irradiation. Oncogene 2009, 28, 1241–1247. [Google Scholar] [CrossRef] [Green Version]
  70. Munshi, A.; Ramesh, R. Mitogen-activated protein kinases and their role in radiation response. Genes Cancer 2013, 4, 401–408. [Google Scholar] [CrossRef]
  71. Broustas, C.G.; Duval, A.J.; Chaudhary, K.R.; Friedman, R.A.; Virk, R.K.; Lieberman, H.B. Targeting MEK5 impairs nonhomologous end-joining repair and sensitizes prostate cancer to DNA damaging agents. Oncogene 2020, 39, 2467–2477. [Google Scholar] [CrossRef]
  72. Jiang, W.; Jin, G.; Cai, F.; Chen, X.; Cao, N.; Zhang, X.; Liu, J.; Chen, F.; Wang, F.; Dong, W.; et al. Extracellular signal-regulated kinase 5 increases radioresistance of lung cancer cells by enhancing the DNA damage response. Exp. Mol. Med. 2019, 51, 1–20. [Google Scholar] [CrossRef]
  73. Dent, P.; Yacoub, A.; Fisher, P.B.; Hagan, M.P.; Grant, S. MAPK pathways in radiation responses. Oncogene 2003, 22, 5885–5896. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Hong, E.-H.; Lee, S.-J.; Kim, J.-S.; Lee, K.-H.; Um, H.-D.; Kim, J.-H.; Kim, S.-J.; Kim, J.-I.; Hwang, S.-G. Ionizing Radiation Induces Cellular Senescence of Articular Chondrocytes via Negative Regulation of SIRT1 by p38 Kinase. J. Biol. Chem. 2010, 285, 1283–1295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Pandey, P.; Raingeaud, J.; Kaneki, M.; Weichselbaum, R.; Davis, R.J.; Kufe, D.; Kharbanda, S. Activation of p38 mitogen-activated protein kinase by c-Abl-dependent and -independent mechanisms. J. Biol. Chem. 1996, 271, 23775–23779. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Pejchal, J.; Novotný, J.; Mařák, V.; Osterreicher, J.; Tichy, A.; Vávrová, J.; Sinkorová, Z.; Zárybnická, L.; Novotná, E.; Chládek, J.; et al. Activation of p38 MAPK and expression of TGF-β1 in rat colon enterocytes after whole body γ-irradiation. Int. J. Radiat. Biol. 2012, 88, 348–358. [Google Scholar] [CrossRef]
  77. Segreto, H.R.C.; Oshima, C.T.F.; Franco, M.F.; Silva, M.R.R.; Egami, M.I.; Teixeira, V.P.C.; Segreto, R.A. Phosphorylation and cytoplasmic localization of MAPK p38 during apoptosis signaling in bone marrow granulocytes of mice irradiated in vivo and the role of amifostine in reducing these effects. Acta Histochem. 2011, 113, 300–307. [Google Scholar] [CrossRef]
  78. Raman, M.; Earnest, S.; Zhang, K.; Zhao, Y.; Cobb, M.H. TAO kinases mediate activation of p38 in response to DNA damage. EMBO J. 2007, 26, 2005–2014. [Google Scholar] [CrossRef]
  79. Sharma, P.K.; Dwarakanath, B.S.; Varshney, R. Radiosensitization by 2-deoxy-D-glucose and 6-aminonicotinamide involves activation of redox sensitive ASK1-JNK/p38MAPK signaling in head and neck cancer cells. Free Radic. Biol. Med. 2012, 53, 1500–1513. [Google Scholar] [CrossRef]
  80. Bulavin, D.V.; Amundson, S.A.; Fornace, A.J. p38 and Chk1 kinases: Different conductors for the G(2)/M checkpoint symphony. Curr. Opin. Genet. Dev. 2002, 12, 92–97. [Google Scholar] [CrossRef]
  81. He, H.; Lin, K.; Zou, C.; Pan, J.; Fu, W.; Zhou, Y.; Lin, H.; Chen, C.; Su, Y. Knockdown of Annexin A2 Enhances Radiosensitivity by Increasing G2/M-Phase Arrest, Apoptosis and Activating the p38 MAPK-HSP27 Pathway in Nasopharyngeal Carcinoma. Front. Oncol. 2022, 12, 769544. [Google Scholar] [CrossRef] [PubMed]
  82. Lafarga, V.; Cuadrado, A.; Lopez de Silanes, I.; Bengoechea, R.; Fernandez-Capetillo, O.; Nebreda, Á.R. p38 Mitogen-activated protein kinase- and HuR-dependent stabilization of p21(Cip1) mRNA mediates the G(1)/S checkpoint. Mol. Cell. Biol. 2009, 29, 4341–4351. [Google Scholar] [CrossRef] [PubMed]
  83. Wang, X.; McGowan, C.H.; Zhao, M.; He, L.; Downey, J.S.; Fearns, C.; Wang, Y.; Huang, S.; Han, J. Involvement of the MKK6-p38gamma cascade in gamma-radiation-induced cell cycle arrest. Mol. Cell. Biol. 2000, 20, 4543–4552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Trempolec, N.; Muñoz, J.P.; Slobodnyuk, K.; Marin, S.; Cascante, M.; Zorzano, A.; Nebreda, Á.R. Induction of oxidative metabolism by the p38α/MK2 pathway. Sci. Rep. 2017, 7, 11367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Tormos, A.M.; Taléns-Visconti, R.; Nebreda, A.R.; Sastre, J. p38 MAPK: A dual role in hepatocyte proliferation through reactive oxygen species. Free Radic. Res. 2013, 47, 905–916. [Google Scholar] [CrossRef] [PubMed]
  86. Lin, F.; Luo, J.; Gao, W.; Wu, J.; Shao, Z.; Wang, Z.; Meng, J.; Ou, Z.; Yang, G. COX-2 promotes breast cancer cell radioresistance via p38/MAPK-mediated cellular anti-apoptosis and invasiveness. Tumour Biol. 2013, 34, 2817–2826. [Google Scholar] [CrossRef]
  87. Tang, F.; Zhang, R.; Wang, J. Cyclooxygenase-2-Mediated Up-Regulation of Mitochondrial Transcription Factor A Mitigates the Radio-Sensitivity of Cancer Cells. Int. J. Mol. Sci. 2019, 20, 1218. [Google Scholar] [CrossRef] [Green Version]
  88. Hur, J.-M.; Yun, H.-J.; Yang, S.-H.; Lee, W.-Y.; Joe, M.-H.; Kim, D. Gliotoxin enhances radiotherapy via inhibition of radiation-induced GADD45a, p38, and NFkappaB activation. J. Cell. Biochem. 2008, 104, 2174–2184. [Google Scholar] [CrossRef]
  89. Choi, C.-H.; Xu, H.; Bark, H.; Lee, T.-B.; Yun, J.; Kang, S.-I.; Oh, Y.-K. Balance of NF-kappaB and p38 MAPK is a determinant of radiosensitivity of the AML-2 and its doxorubicin-resistant cell lines. Leuk. Res. 2007, 31, 1267–1276. [Google Scholar] [CrossRef]
  90. Viktorsson, K.; Ekedahl, J.; Lindebro, M.C.; Lewensohn, R.; Zhivotovsky, B.; Linder, S.; Shoshan, M.C. Defective stress kinase and Bak activation in response to ionizing radiation but not cisplatin in a non-small cell lung carcinoma cell line. Exp. Cell Res. 2003, 289, 256–264. [Google Scholar] [CrossRef]
  91. Koturbash, I.; Merrifield, M.; Kovalchuk, O. Fractionated exposure to low doses of ionizing radiation results in accumulation of DNA damage in mouse spleen tissue and activation of apoptosis in a p53/Atm-independent manner. Int. J. Radiat. Biol. 2017, 93, 148–155. [Google Scholar] [CrossRef] [PubMed]
  92. Kumar, P.; Miller, A.I.; Polverini, P.J. p38 MAPK mediates gamma-irradiation-induced endothelial cell apoptosis, and vascular endothelial growth factor protects endothelial cells through the phosphoinositide 3-kinase-Akt-Bcl-2 pathway. J. Biol. Chem. 2004, 279, 43352–43360. [Google Scholar] [CrossRef] [PubMed]
  93. Choi, S.-Y.; Kim, M.-J.; Kang, C.-M.; Bae, S.; Cho, C.-K.; Soh, J.-W.; Kim, J.-H.; Kang, S.; Chung, H.Y.; Lee, Y.-S.; et al. Activation of Bak and Bax through c-abl-protein kinase Cdelta-p38 MAPK signaling in response to ionizing radiation in human non-small cell lung cancer cells. J. Biol. Chem. 2006, 281, 7049–7059. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Ding, W.; Lu, Y.; Zhou, A.; Chen, Y.; Wang, Z.; Wang, L.; Tian, Y. Mast1 mediates radiation-induced gastric injury via the P38 MAPK pathway. Exp. Cell Res. 2021, 409, 112913. [Google Scholar] [CrossRef]
  95. Salim, H.; Akbar, N.S.; Zong, D.; Vaculova, A.H.; Lewensohn, R.; Moshfegh, A.; Viktorsson, K.; Zhivotovsky, B. miRNA-214 modulates radiotherapy response of non-small cell lung cancer cells through regulation of p38MAPK, apoptosis and senescence. Br. J. Cancer 2012, 107, 1361–1373. [Google Scholar] [CrossRef] [Green Version]
  96. Kim, M.-J.; Choi, S.-Y.; Park, I.-C.; Hwang, S.-G.; Kim, C.; Choi, Y.-H.; Kim, H.; Lee, K.-H.; Lee, S.-J. Opposing roles of c-Jun NH2-terminal kinase and p38 mitogen-activated protein kinase in the cellular response to ionizing radiation in human cervical cancer cells. Mol. Cancer Res. 2008, 6, 1718–1731. [Google Scholar] [CrossRef] [Green Version]
  97. Ståhl, S.; Fung, E.; Adams, C.; Lengqvist, J.; Mörk, B.; Stenerlöw, B.; Lewensohn, R.; Lehtiö, J.; Zubarev, R.; Viktorsson, K. Proteomics and pathway analysis identifies JNK signaling as critical for high linear energy transfer radiation-induced apoptosis in non-small lung cancer cells. Mol. Cell. Proteom. MCP 2009, 8, 1117–1129. [Google Scholar] [CrossRef] [Green Version]
  98. Whitaker, R.H.; Cook, J.G. Stress Relief Techniques: p38 MAPK Determines the Balance of Cell Cycle and Apoptosis Pathways. Biomolecules 2021, 11, 1444. [Google Scholar] [CrossRef]
  99. Al-Jumayli, M.; Brown, S.L.; Chetty, I.J.; Extermann, M.; Movsas, B. The Biological Process of Aging and the Impact of Ionizing Radiation. Semin. Radiat. Oncol. 2022, 32, 172–178. [Google Scholar] [CrossRef]
  100. Han, J.; Sun, P. The pathways to tumor suppression via route p38. Trends Biochem. Sci. 2007, 32, 364–371. [Google Scholar] [CrossRef]
  101. Wang, Y.; Scheiber, M.N.; Neumann, C.; Calin, G.A.; Zhou, D. MicroRNA regulation of ionizing radiation-induced premature senescence. Int. J. Radiat. Oncol. Biol. Phys. 2011, 81, 839–848. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Fernández-Aroca, D.M.; García-Flores, N.; Frost, S.; Jiménez-Suarez, J.; Rodríguez-González, A.; Fernández-Aroca, P.; Sabater, S.; Andrés, I.; Garnés-García, C.; Belandia, B.; et al. Genetic perturbation of MAPK11 (p38β) promotes radiosensitivity by enhancing IR-associated senescence. bioRxiv 2022. [Google Scholar] [CrossRef]
  103. Roy, A.; Bera, S.; Saso, L.; Dwarakanath, B.S. Role of autophagy in tumor response to radiation: Implications for improving radiotherapy. Front. Oncol. 2022, 12, 957373. [Google Scholar] [CrossRef] [PubMed]
  104. de la Cruz-Morcillo, M.A.; Valero, M.L.L.; Callejas-Valera, J.L.; Arias-González, L.; Melgar-Rojas, P.; Galán-Moya, E.M.; García-Gil, E.; García-Cano, J.; Sánchez-Prieto, R. P38MAPK is a major determinant of the balance between apoptosis and autophagy triggered by 5-fluorouracil: Implication in resistance. Oncogene 2011, 31, 1073–1085. [Google Scholar] [CrossRef]
  105. Králová, V.; Benešová, S.; Cervinka, M.; Rudolf, E. Selenite-induced apoptosis and autophagy in colon cancer cells. Toxicol. Vitr. Int. J. Publ. Assoc. BIBRA 2012, 26, 258–268. [Google Scholar] [CrossRef]
  106. Zhu, Q.; Guo, Y.; Chen, S.; Fu, D.; Li, Y.; Li, Z.; Ni, C. Irinotecan Induces Autophagy-Dependent Apoptosis and Positively Regulates ROS-Related JNK- and P38-MAPK Pathways in Gastric Cancer Cells. OTT 2020, 13, 2807–2817. [Google Scholar] [CrossRef] [Green Version]
  107. Qiao, L.; Chen, Y.; Liang, N.; Xie, J.; Deng, G.; Chen, F.; Wang, X.; Liu, F.; Li, Y.; Zhang, J. Targeting Epithelial-to-Mesenchymal Transition in Radioresistance: Crosslinked Mechanisms and Strategies. Front. Oncol. 2022, 12, 775238. [Google Scholar] [CrossRef]
  108. Jung, J.-W.; Hwang, S.-Y.; Hwang, J.-S.; Oh, E.-S.; Park, S.; Han, I.-O. Ionising radiation induces changes associated with epithelial-mesenchymal transdifferentiation and increased cell motility of A549 lung epithelial cells. Eur. J. Cancer 2007, 43, 1214–1224. [Google Scholar] [CrossRef]
  109. Pan, Y.; Zhou, C.; Yuan, D.; Zhang, J.; Shao, C. Radiation Exposure Promotes Hepatocarcinoma Cell Invasion through Epithelial Mesenchymal Transition Mediated by H2S/CSE Pathway. Radiat. Res. 2016, 185, 96–105. [Google Scholar] [CrossRef]
  110. Berggren, K.L.; Restrepo Cruz, S.; Hixon, M.D.; Cowan, A.T.; Keysar, S.B.; Craig, S.; James, J.; Barry, M.; Ozbun, M.A.; Jimeno, A.; et al. MAPKAPK2 (MK2) inhibition mediates radiation-induced inflammatory cytokine production and tumor growth in head and neck squamous cell carcinoma. Oncogene 2019, 38, 7329–7341. [Google Scholar] [CrossRef]
  111. Chowdhury, P.; Dey, P.; Ghosh, S.; Sarma, A.; Ghosh, U. Reduction of metastatic potential by inhibiting EGFR/Akt/p38/ERK signaling pathway and epithelial-mesenchymal transition after carbon ion exposure is potentiated by PARP-1 inhibition in non-small-cell lung cancer. BMC Cancer 2019, 19, 829. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Chowdhury, P.; Dey, P.; De, D.; Ghosh, U. Gamma ray-induced in vitro cell migration via EGFR/ERK/Akt/p38 activation is prevented by olaparib pretreatment. Int. J. Radiat. Biol. 2020, 96, 651–660. [Google Scholar] [CrossRef] [PubMed]
  113. Sharma, A.K.; Khan, S.A.; Sharda, A.; Reddy, D.V.; Gupta, S. MKP1 phosphatase mediates G1-specific dephosphorylation of H3Serine10P in response to DNA damage. Mutat. Res. 2015, 778, 71–79. [Google Scholar] [CrossRef] [PubMed]
  114. Candas, D.; Lu, C.-L.; Fan, M.; Chuang, F.Y.S.; Sweeney, C.; Borowsky, A.D.; Li, J.J. Mitochondrial MKP1 is a target for therapy-resistant HER2-positive breast cancer cells. Cancer Res. 2014, 74, 7498–7509. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Jones, R.M.; Sloane, V.M.; Wu, H.; Luo, L.; Kumar, A.; Kumar, M.V.; Gewirtz, A.T.; Neish, A.S. Flagellin administration protects gut mucosal tissue from irradiation-induced apoptosis via MKP-7 activity. Gut 2011, 60, 648–657. [Google Scholar] [CrossRef]
  116. Palanichamy, K.; Kanji, S.; Gordon, N.; Thirumoorthy, K.; Jacob, J.R.; Litzenberg, K.T.; Patel, D.; Chakravarti, A. NNMT Silencing Activates Tumor Suppressor PP2A, Inactivates Oncogenic STKs, and Inhibits Tumor Forming Ability. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2017, 23, 2325–2334. [Google Scholar] [CrossRef] [Green Version]
  117. Hong, C.S.; Ho, W.; Zhang, C.; Yang, C.; Elder, J.B.; Zhuang, Z. LB100, a small molecule inhibitor of PP2A with potent chemo- and radio-sensitizing potential. Cancer Biol. Ther. 2015, 16, 821–833. [Google Scholar] [CrossRef] [Green Version]
  118. Taher, M.M.; Hershey, C.M.; Oakley, J.D.; Valerie, K. Role of the p38 and MEK-1/2/p42/44 MAP kinase pathways in the differential activation of human immunodeficiency virus gene expression by ultraviolet and ionizing radiation. Photochem. Photobiol. 2000, 71, 455–459. [Google Scholar] [CrossRef]
  119. Choi, J.-A.; Park, M.-T.; Kang, C.-M.; Um, H.-D.; Bae, S.; Lee, K.-H.; Kim, T.-H.; Kim, J.-H.; Cho, C.-K.; Lee, Y.-S.; et al. Opposite effects of Ha-Ras and Ki-Ras on radiation-induced apoptosis via differential activation of PI3K/Akt and Rac/p38 mitogen-activated protein kinase signaling pathways. Oncogene 2004, 23, 9–20. [Google Scholar] [CrossRef] [Green Version]
  120. Pal, S.; Yadav, P.; Sainis, K.B.; Shankar, B.S. TNF-α and IGF-1 differentially modulate ionizing radiation responses of lung cancer cell lines. Cytokine 2018, 101, 89–98. [Google Scholar] [CrossRef]
  121. Sproull, M.; Shankavaram, U.; Camphausen, K. Novel Murine Biomarkers of Radiation Exposure Using An Aptamer-Based Proteomic Technology. Front. Pharmacol. 2021, 12, 633131. [Google Scholar] [CrossRef] [PubMed]
  122. Abbasi, A.N.; Hammad Khan, A.M.; Mansha, M.A.; Shaukat, F.; Tahseen, R. Abscopal Effect: An Old Concept with a New Horizon. Gulf J. Oncol. 2021, 1, 60–66. [Google Scholar]
  123. Ghosh, S.; Kumar, A.; Tripathi, R.P.; Chandna, S. Connexin-43 regulates p38-mediated cell migration and invasion induced selectively in tumour cells by low doses of γ-radiation in an ERK-1/2-independent manner. Carcinogenesis 2014, 35, 383–395. [Google Scholar] [CrossRef] [PubMed]
  124. Asur, R.; Balasubramaniam, M.; Marples, B.; Thomas, R.A.; Tucker, J.D. Involvement of MAPK proteins in bystander effects induced by chemicals and ionizing radiation. Mutat. Res. 2010, 686, 15–29. [Google Scholar] [CrossRef]
  125. Hu, S.; Zhu, L.; Song, Y.; Zhao, X.; Chen, Q.; Pan, Y.; Zhang, J.; Bai, Y.; Zhang, H.; Shao, C. Radiation-induced abscopal reproductive effect is driven by TNF-α/p38 MAPK/Rac1 axis in Sertoli cells. Theranostics 2021, 11, 5742–5758. [Google Scholar] [CrossRef]
  126. Fan, Z.; Han, Y.; Ye, Y.; Liu, C.; Cai, H. l-carnitine preserves cardiac function by activating p38 MAPK/Nrf2 signalling in hearts exposed to irradiation. Eur. J. Pharmacol. 2017, 804, 7–12. [Google Scholar] [CrossRef]
  127. Song, Y.-H.; Chai, Q.; Wang, N.-L.; Yang, F.-F.; Wang, G.-H.; Hu, J.-Y. X-rays induced IL-8 production in lung cancer cells via p38/MAPK and NF-κB pathway. Int. J. Radiat. Biol. 2020, 96, 1374–1381. [Google Scholar] [CrossRef]
  128. Sundaramoorthy, P.; Wang, Q.; Zheng, Z.; Jiao, Y.; Chen, B.J.; Doan, P.L.; Chao, N.J.; Kang, Y. Thioredoxin mitigates radiation-induced hematopoietic stem cell injury in mice. Stem Cell Res. Ther. 2017, 8, 263. [Google Scholar] [CrossRef] [Green Version]
  129. Lu, L.; Wang, Y.-Y.; Zhang, J.-L.; Li, D.-G.; Meng, A.-M. p38 MAPK Inhibitor Insufficiently Attenuates HSC Senescence Administered Long-Term after 6 Gy Total Body Irradiation in Mice. Int. J. Mol. Sci. 2016, 17, 905. [Google Scholar] [CrossRef] [Green Version]
  130. Li, D.; Wang, Y.; Wu, H.; Lu, L.; Zhang, H.; Chang, J.; Zhai, Z.; Zhang, J.; Wang, Y.; Zhou, D.; et al. Mitigation of ionizing radiation-induced bone marrow suppression by p38 inhibition and G-CSF administration. J. Radiat. Res. 2011, 52, 712–716. [Google Scholar] [CrossRef] [Green Version]
  131. Konkol, M.; Śniatała, P.; Milecki, P. Radiation-induced lung injury-what do we know in the era of modern radiotherapy? Rep. Pract. Oncol. Radiother. J. Greatpoland Cancer Cent. Pozn. Pol. Soc. Radiat. Oncol. 2022, 27, 552–565. [Google Scholar] [CrossRef]
  132. Yahyapour, R.; Amini, P.; Rezapoor, S.; Rezaeyan, A.; Farhood, B.; Cheki, M.; Fallah, H.; Najafi, M. Targeting of Inflammation for Radiation Protection and Mitigation. Curr. Mol. Pharmacol. 2018, 11, 203–210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Arora, A.; Bhuria, V.; Singh, S.; Pathak, U.; Mathur, S.; Hazari, P.P.; Roy, B.G.; Sandhir, R.; Soni, R.; Dwarakanath, B.S.; et al. Amifostine analog, DRDE-30, alleviates radiation induced lung damage by attenuating inflammation and fibrosis. Life Sci. 2022, 298, 120518. [Google Scholar] [CrossRef] [PubMed]
  134. Lu, L.; Li, W.; Chen, L.; Su, Q.; Wang, Y.; Guo, Z.; Lu, Y.; Liu, B.; Qin, S. Radiation-induced intestinal damage: Latest molecular and clinical developments. Future Oncol. 2019, 15, 4105–4118. [Google Scholar] [CrossRef] [PubMed]
  135. Meena, S.K.; Joriya, P.R.; Yadav, S.M.; Kumar, R.; Meena, P.; Patel, D.D. Modulation of radiation-induced intestinal injury by radioprotective agents: A cellular and molecular perspectives. Rev. Environ. Health 2022. [Google Scholar] [CrossRef] [PubMed]
  136. Tanoue, T.; Yamamoto, T.; Maeda, R.; Nishida, E. A Novel MAPK phosphatase MKP-7 acts preferentially on JNK/SAPK and p38 alpha and beta MAPKs. J. Biol. Chem. 2001, 276, 26629–26639. [Google Scholar] [CrossRef] [Green Version]
  137. Sha, H.; Gu, Y.; Shen, W.; Zhang, L.; Qian, F.; Zhao, Y.; Li, H.; Zhang, T.; Lu, W. Rheinic acid ameliorates radiation-induced acute enteritis in rats through PPAR-γ/NF-κB. Genes Genom. 2019, 41, 909–917. [Google Scholar] [CrossRef]
  138. Kiang, J.G.; Cannon, G.; Olson, M.G.; Smith, J.T.; Anderson, M.N.; Zhai, M.; Umali, M.V.; Ho, K.; Ho, C.; Cui, W.; et al. Female Mice are More Resistant to the Mixed-Field (67% Neutron + 33% Gamma) Radiation-Induced Injury in Bone Marrow and Small Intestine than Male Mice due to Sustained Increases in G-CSF and the Bcl-2/Bax Ratio and Lower miR-34a and MAPK Activation. Radiat. Res. 2022, 198, 120–133. [Google Scholar] [CrossRef]
  139. de la Cruz-Morcillo, M.A.; García-Cano, J.; Arias-González, L.; García-Gil, E.; Artacho-Cordón, F.; Ríos-Arrabal, S.; Valero, M.L.; Cimas, F.J.; Serrano-Oviedo, L.; Villas, M.V.; et al. Abrogation of the p38 MAPK α signaling pathway does not promote radioresistance but its activity is required for 5-Fluorouracil-associated radiosensitivity. Cancer Lett. 2013, 335, 66–74. [Google Scholar] [CrossRef]
  140. Wang, Y.; Li, Y.; Yang, L.; Yin, D. Intermittent low dose irradiation enhances the effectiveness of radio- and chemo-therapy for human colorectal adenocarcinoma cell line HT-29. Oncol. Rep. 2017, 38, 591–597. [Google Scholar] [CrossRef] [Green Version]
  141. Martinez-Useros, J.; Moreno, I.; Fernandez-Aceñero, M.J.; Rodriguez-Remirez, M.; Borrero-Palacios, A.; Cebrian, A.; Gomez del Pulgar, T.; del Puerto-Nevado, L.; Li, W.; Puime-Otin, A.; et al. The potential predictive value of DEK expression for neoadjuvant chemoradiotherapy response in locally advanced rectal cancer. BMC Cancer 2018, 18, 144. [Google Scholar] [CrossRef] [Green Version]
  142. Pascual-Serra, R.; Fernández-Aroca, D.M.; Sabater, S.; Roche, O.; Andrés, I.; Ortega-Muelas, M.; Arconada-Luque, E.; García-Flores, N.; Bossi, G.; Belandia, B.; et al. p38β (MAPK11) mediates gemcitabine-associated radiosensitivity in sarcoma experimental models. Radiother. Oncol. 2021, 156, 136–144. [Google Scholar] [CrossRef]
  143. Ho, S.-Y.; Wu, W.-J.; Chiu, H.-W.; Chen, Y.-A.; Ho, Y.-S.; Guo, H.-R.; Wang, Y.-J. Arsenic trioxide and radiation enhance apoptotic effects in HL-60 cells through increased ROS generation and regulation of JNK and p38 MAPK signaling pathways. Chem.-Biol. Interact. 2011, 193, 162–171. [Google Scholar] [CrossRef]
  144. Kale, Ş.; Korcum, A.F.; Dündar, E.; Erin, N. HSP90 inhibitor PU-H71 increases radiosensitivity of breast cancer cells metastasized to visceral organs and alters the levels of inflammatory mediators. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2020, 393, 253–262. [Google Scholar] [CrossRef]
  145. Hur, J.-M.; Hyun, M.-S.; Lim, S.-Y.; Lee, W.-Y.; Kim, D. The combination of berberine and irradiation enhances anti-cancer effects via activation of p38 MAPK pathway and ROS generation in human hepatoma cells. J. Cell. Biochem. 2009, 107, 955–964. [Google Scholar] [CrossRef] [PubMed]
  146. Bernardo, P.S.; Guimarães, G.H.C.; De Faria, F.C.C.; Longo, G.M.D.C.; Lopes, G.P.D.F.; Netto, C.D.; Costa, P.R.R.; Maia, R.C. LQB-118 compound inhibits migration and induces cell death in glioblastoma cells. Oncol. Rep. 2020, 43, 346–357. [Google Scholar] [CrossRef] [PubMed]
  147. Cho, H.-J.; Ahn, K.-C.; Choi, J.Y.; Hwang, S.-G.; Kim, W.-J.; Um, H.-D.; Park, J.K. Luteolin acts as a radiosensitizer in non-small cell lung cancer cells by enhancing apoptotic cell death through activation of a p38/ROS/caspase cascade. Int. J. Oncol. 2015, 46, 1149–1158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Yuan, S.; Qiao, T.; Li, X.; Zhuang, X.; Chen, W.; Chen, X.; Zhang, Q. Toll-like receptor 9 activation by CpG oligodeoxynucleotide 7909 enhances the radiosensitivity of A549 lung cancer cells via the p53 signaling pathway. Oncol. Lett. 2018, 15, 5271–5279. [Google Scholar] [CrossRef] [Green Version]
  149. Cheol Son, J.; Woo Kang, D.; Mo Yang, K.; Choi, K.-Y.; Gen Son, T.; Min, D.S. Phospholipase D inhibitor enhances radiosensitivity of breast cancer cells. Exp. Mol. Med. 2013, 45, e38. [Google Scholar] [CrossRef]
  150. Camero, S.; Vitali, G.; Pontecorvi, P.; Ceccarelli, S.; Anastasiadou, E.; Cicchetti, F.; Flex, E.; Pomella, S.; Cassandri, M.; Rota, R.; et al. DNMT3A and DNMT3B Targeting as an Effective Radiosensitizing Strategy in Embryonal Rhabdomyosarcoma. Cells 2021, 10, 2956. [Google Scholar] [CrossRef]
  151. Wang, K.; Tang, J.; Liu, X.; Wang, Y.; Chen, W.; Zheng, R. UBR5 regulates proliferation and radiosensitivity in human laryngeal carcinoma via the p38/MAPK signaling pathway. Oncol. Rep. 2020, 44, 685–697. [Google Scholar] [CrossRef] [PubMed]
  152. Zhou, X.; Zhang, W.; Dou, M.; Li, Z.; Liu, Z.; Li, J.; Tian, C.; Yao, Y.; Wang, C.; Li, Y.; et al. 125I seeds inhibit proliferation and promote apoptosis in cholangiocarcinoma cells by regulating the AGR2-mediated p38 MAPK pathway. Cancer Lett. 2022, 524, 29–41. [Google Scholar] [CrossRef] [PubMed]
  153. Tang, T.; Wang, L.-X.; Yang, M.-L.; Zhang, R.-M. lncRNA TPTEP1 inhibits stemness and radioresistance of glioma through miR-106a-5p-mediated P38 MAPK signaling. Mol. Med. Rep. 2020, 22, 4857–4867. [Google Scholar] [CrossRef] [PubMed]
  154. Yang, W.; Shen, Y.; Wei, J.; Liu, F. MicroRNA-153/Nrf-2/GPx1 pathway regulates radiosensitivity and stemness of glioma stem cells via reactive oxygen species. Oncotarget 2015, 6, 22006–22027. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Biau, J.; Thivat, E.; Chautard, E.; Stefan, D.; Boone, M.; Chauffert, B.; Bourgne, C.; Richard, D.; Molnar, I.; Levesque, S.; et al. Phase 1 trial of ralimetinib (LY2228820) with radiotherapy plus concomitant temozolomide in the treatment of newly diagnosed glioblastoma. Radiother. Oncol. 2021, 154, 227–234. [Google Scholar] [CrossRef]
  156. Denny, W.A. Inhibitors and Activators of the p38 Mitogen-Activated MAP Kinase (MAPK) Family as Drugs to Treat Cancer and Inflammation. Curr. Cancer Drug Targets 2022, 22, 209–220. [Google Scholar] [CrossRef]
  157. Donoghue, C.; Cubillos-Rojas, M.; Gutierrez-Prat, N.; Sanchez-Zarzalejo, C.; Verdaguer, X.; Riera, A.; Nebreda, Á.R. Optimal linker length for small molecule PROTACs that selectively target p38α and p38β for degradation. Eur. J. Med. Chem. 2020, 201, 112451. [Google Scholar] [CrossRef]
Figure 1. p38 MAPK diversity in structure and activation. (a) Diagram of the four human p38 kinases, in-dicating amino acid numbers and the different domains: The CD-charged docking domain is a negatively charged region involved in high-affinity docking interactions with positively charged substrates. The ED domain contributes to substrate docking. The ATP binding site and the phos-phorylated Thr and Tyr residues of the activation loop are also indicated. The PDZ binding site is a protein–protein interaction motif exclusive to p38γ. (b) Diagram of the MAP2Ks and their speci-ficity for the different p38MAPKs.
Figure 1. p38 MAPK diversity in structure and activation. (a) Diagram of the four human p38 kinases, in-dicating amino acid numbers and the different domains: The CD-charged docking domain is a negatively charged region involved in high-affinity docking interactions with positively charged substrates. The ED domain contributes to substrate docking. The ATP binding site and the phos-phorylated Thr and Tyr residues of the activation loop are also indicated. The PDZ binding site is a protein–protein interaction motif exclusive to p38γ. (b) Diagram of the MAP2Ks and their speci-ficity for the different p38MAPKs.
Cancers 15 00861 g001
Figure 2. p38α controls cell cycle arrest in G1 through several mechanisms. Within its family, p38α is the main regulator of the G1/S checkpoint through different mechanisms, implicating key proteins, such as pRB, p53 and p21/WAF, in this checkpoint.
Figure 2. p38α controls cell cycle arrest in G1 through several mechanisms. Within its family, p38α is the main regulator of the G1/S checkpoint through different mechanisms, implicating key proteins, such as pRB, p53 and p21/WAF, in this checkpoint.
Cancers 15 00861 g002
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

García-Flores, N.; Jiménez-Suárez, J.; Garnés-García, C.; Fernández-Aroca, D.M.; Sabater, S.; Andrés, I.; Fernández-Aramburo, A.; Ruiz-Hidalgo, M.J.; Belandia, B.; Sanchez-Prieto, R.; et al. P38 MAPK and Radiotherapy: Foes or Friends? Cancers 2023, 15, 861. https://doi.org/10.3390/cancers15030861

AMA Style

García-Flores N, Jiménez-Suárez J, Garnés-García C, Fernández-Aroca DM, Sabater S, Andrés I, Fernández-Aramburo A, Ruiz-Hidalgo MJ, Belandia B, Sanchez-Prieto R, et al. P38 MAPK and Radiotherapy: Foes or Friends? Cancers. 2023; 15(3):861. https://doi.org/10.3390/cancers15030861

Chicago/Turabian Style

García-Flores, Natalia, Jaime Jiménez-Suárez, Cristina Garnés-García, Diego M. Fernández-Aroca, Sebastia Sabater, Ignacio Andrés, Antonio Fernández-Aramburo, María José Ruiz-Hidalgo, Borja Belandia, Ricardo Sanchez-Prieto, and et al. 2023. "P38 MAPK and Radiotherapy: Foes or Friends?" Cancers 15, no. 3: 861. https://doi.org/10.3390/cancers15030861

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