IMR Press / FBL / Volume 27 / Issue 5 / DOI: 10.31083/j.fbl2705167
Open Access Review
MAP3K Family Review and Correlations with Patient Survival Outcomes in Various Cancer Types
Show Less
1 Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
2 Department of Pharmacological Sciences, Division of Medicinal Chemistry, Mylan School of Pharmacy, Duquesne University, Pittsburgh, PA 15218, USA
3 Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
4 UNC Eshelman School of Pharmacy and UNC Lineberger Comprehensive Cancer Center, Chemical Biology and Medicinal Chemistry Division, SGC-UNC, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
*Correspondence: knguyen11@tulane.edu (Khoa Nguyen); mburow@tulane.edu (Matthew E. Burow)
These authors contributed equally.
Academic Editors: Guan Chen and Graham Pawelec
Front. Biosci. (Landmark Ed) 2022, 27(5), 167; https://doi.org/10.31083/j.fbl2705167
Submitted: 1 January 2022 | Revised: 14 February 2022 | Accepted: 24 February 2022 | Published: 23 May 2022
(This article belongs to the Special Issue MAPKs and Oncogenesis)
Copyright: © 2022 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

The mitogen-activated protein kinase (MAPK) pathways are ubiquitous in cellular signaling and are essential for proper biological functions. Disruptions in this signaling axis can lead to diseases such as the development of cancer. In this review, we discuss members of the MAP3K family and correlate their mRNA expression levels to patient survival outcomes in different cancers. Furthermore, we highlight the importance of studying the MAP3K family due to their important roles in the larger, overall MAPK pathway, relationships with cancer progression, and the understudied status of these kinases.

Keywords
MAP kinases
MAPK
MAP3K
understudied kinase
Dark Kinome
Illuminating the Druggable Genome
cancer
1. Introduction

Mitogen-activated protein kinase (MAPK) pathways are crucial signaling networks that link extracellular signals to machinery that controls cellular processes such as growth, proliferation, differentiation, migration, and apoptosis. Once activated by a stimulus, MAPK pathways are characterized by three sequential phosphorylation of MAPK kinase kinases (MAP3K, MEK kinases, or MKKKs) to MAPK kinases (MAP2K, MEK, or MKKs) to MAPKs. The 4 well-known and conventional MAPK subfamilies are extracellular signal-regulated kinases 1 and 2 (ERK1/2), c- Jun amino-terminal kinases 1 to 3 (JNK1 to 3), p38 (α, β, γ, and δ), and ERK5 families [1].

MAP3Ks are serine/threonine kinases that act upstream of MAP2Ks and MAPKs. There are 24 characterized MAP3Ks, named from MAP3K1 to MAP3K21 plus B-Raf, C-Raf, and A-Raf [2].

Supplementary Table 1 contains the Uniprot number, synonyms, chromosome location, and HGNC ID for each MAP3K isoform. It has been well-described that MAPK signaling pathways can be dysregulated and some MAP3Ks become mutated in cancers [3]. Among the aforementioned MAP3Ks, the role of B-Raf in cancers is the most thoroughly characterized. Activating mutations in the BRAF oncogenes resulting in constitutive activation of MEK1/2 and subsequent activation of ERK1/2 are seen in some 70% of melanomas, some 10% of colorectal cancers, and some 30–70% of papillary thyroid carcinomas [4, 5, 6, 7]. Supplementary Table 2 contains a compilation of MAPK compound inhibitors and their investigations in different diseases, mechanisms of action, and stage of study [8]. However, except for B-Raf, the role of other MAP3Ks in cancers has not been fully investigated.

This review seeks to address the gap in knowledge of MAP3K family members and establish a foundation for elucidating functions of novel MAP3Ks. We approach this by discussing existing literature and by determining the correlations between the mRNA expression of 21 MAP3K isoforms and patient survival across 21 cancer types using Kaplan-Meier online plotter. Additionally, we also use the Pharos user interface to the Knowledge Management Center’s (KMC) Illuminating the Druggable Genome (IDG) program funded by the National Institutes of Health (NIH) Common Fund to quantify the novelty of each MAP3K family member by assigning them each a PubMed and Novelty score. Lastly, we reference the NIH FOA RFA-RM-21-012, titled Pilot Projects Investigating Understudied G Protein-Coupled Receptors, Ion Channels, and Protein Kinases, to emphasize the importance and opportunities associated with studying these kinase family members. IDG-eligible kinases are prioritized by this funding opportunity to support the generation of data and tools to study understudied kinases.

2. Method
2.1 The KM Plotter Online Tool was Used to Determine the mRNA Expression of MAP3Ks and Patient Survival

The KM plotter online tool was used to investigate the mRNA expression of 21 MAP3K isoforms and patient survival across 21 cancer types: bladder carcinoma, breast cancer, cervical squamous cell carcinoma, esophageal adenocarcinoma, esophageal squamous cell carcinoma, head-neck squamous cell carcinoma, kidney renal clear cell carcinoma, kidney renal papillary cell carcinoma, liver hepatocellular carcinoma, lung adenocarcinoma, lung squamous cell carcinoma, ovarian cancer, pancreatic ductal adenocarcinoma, pheochromocytoma and paraganglioma, rectum adenocarcinoma, sarcoma, stomach adenocarcinoma, testicular germ cell tumor, thymoma, thyroid carcinoma, and uterine corpus endometrial carcinoma [9, 10]. Parameters for cutoffs were p < 0.05 and Hazard Ratios excluding a value equal to 1.0. The results section of this manuscript and primary tables only discusses and displays data that are statistically significant. For complete analysis, including results with insignificant p-values, refer to Supplementary Table 3.

2.2 PubMed and Novelty Score Analysis

PubMed and Novelty scores were generated through Pharos (Pharos.NIH.gov), a user interface for the Knowledge Management Center (KMC) for the Illuminating the Druggable Genome (IDG) program funded by the National Institutes of Health (NIH) Common Fund. The PubMed score is described as: “Jensen Lab generated fractional counting score for the prevalence of this target in PubMed articles”. The Novelty score is described as: “Tin-X metric for the relative scarcity of specific publications for this target”. To summarize score interpretations, higher PubMed scores represent higher availability of publications and lower log Novelty scores (more negative) represent lower novelty for the kinase of interest based on the number of PubMed articles. MAPK1 (ERK1/2) is provided as an exemplar of a well-characterized kinase. Additionally, MAP3K members (MAP3K10, MAP3K14, MAP3K15, MAP3K16, MAP3K17, and MAP3K21) described as “2021 NIH designated understudied kinase(s)” are based on the funding announcement RFA- RM-21-012, titled Pilot Projects Investigating Understudied G Protein-Coupled Receptors, Ion Channels, and Protein Kinases.

3. Result
3.1 MAP3K1A

MAP3K1 is one of the most-studied MAP3K family members with a PubMed Score of 217.17 and a Novelty Score of –5.49 (Table 1). Unique among MAP3Ks, only MAP3K1 contains both a kinase domain and a plant homeodomain (PHD) motif, allowing it to regulate downstream protein phosphorylation as well as exhibit E3 ubiquitin ligase activity [11, 12]. MAP3K1 also has a cleavage site that generates a kinase-domain fragment when cleaved by caspase 3, increasing apoptotic response [13]. Therefore, MAP3K1 promotes cell survival or induces apoptosis via an ERK/NF-κB or caspase 3 mechanism, respectively [14, 15]. Consequently, mutations in both pro- survival and pro-apoptotic pathways of MAP3K1 have been identified in cancer [12].

Table 1.PubMed and Novelty scores of MAP3K isoforms.
MAP3K isoforms PubMed score Novelty score (log)
MAPK1 1463.91 –7.4
MAP3K1 217.17 –5.49
MAP3K2 83.41 –4.42
MAP3K3 74.03 –4.13
MAP3K4 43.58 –3.82
MAP3K5 812.85 –6.54
MAP3K6 27.05 –3.29
MAP3K7 209.21 –5.22
MAP3K8 220.14 –5.15
MAP3K9 43.94 –3.72
MAP3K10 25.78 –3.34
MAP3K11 117.01 –4.5
MAP3K12 157.61 –5.07
MAP3K13 200.04 –5.38
MAP3K14 29.23 –3.35
MAP3K15 10.11 –2.11
MAP3K16 57.97 –4.07
MAP3K17 46.59 –3.76
MAP3K18 50.8 –3.88
MAP3K19 4.83 –1.31
MAP3K20 16.1 –2.79
MAP3K21 11.8 –1.96
MAP3K isoforms that are IDG-eligible kinases are in bold italic. MAPK1 is included as an example of a well-characterized kinase.

Mutations in MAP3K1 have been implicated in cancers of breast, prostate, stomach, and diffuse large B cell lymphoma [14]. Among these cancer types, MAP3K1 mutations in breast cancers are most well-studied. Genomic studies revealed MAP3K1 as the second most frequently mutated gene with inactivating mutations in MAP3K1 and MAP2K4 as well in upstream kinases of c-Jun N-terminal kinase (JNK) in the apoptotic pathway identified in luminal A subtype tumors [15, 16]. Moreover, MAP3K1 has been suggested to have a tumor suppressor role in the crosstalk between PI3Kα and MAP3K1 pathways in PIK3CA-mutated luminal/ER+ breast cancers [15]. In glioblastoma, an increase in MAP3K1 was associated with survival of glioma, therapeutic resistance to temozolomide chemotherapy, and radiotherapy [17]. Suppression of MAP3K1- mediated androgen receptor (AR)-dependent apoptosis could lead to chemotherapy resistance in AR+ prostate cancer [18].

The mRNA expression of MAP3K1 was associated with increased survival in cervical squamous cell carcinoma, esophageal squamous cell carcinoma, head-neck squamous cell carcinoma, kidney renal clear cell carcinoma, lung adenocarcinoma, stomach adenocarcinoma, and thyroid carcinoma. MAP3K1 expression was negatively associated with patient survival in kidney renal papillary cell carcinoma, liver hepatocellular carcinoma, and pancreatic ductal adenocarcinoma (Table 2). MAP3K1 mRNA expression in other cancer types did not make the statistical cutoff.

Table 2.Hazard Ratios (HR) correlating patient survival and mRNA expression of MAP3K isoforms using Kaplan-Meier plotter.
MAP3K isoforms Cancer type HR p value
MAP3K1 Cervical squamous cell carcinoma 0.57 (0.36–0.91) 0.017
Esophageal Squamous Cell Carcinoma 0.34 (0.15–0.78) 0.008
Head-neck squamous cell carcinoma 0.62 (0.44–0.87) 0.0055
Kidney renal clear cell carcinoma 0.54 (0.4–0.73) 0.000042
Lung adenocarcinoma 0.55 (0.38–0.81) 0.0019
Stomach adenocarcinoma 0.68 (0.48–0.97) 0.03
Thyroid carcinoma 0.18 (0.04–0.78) 0.01
Kidney renal papillary cell carcinoma 2.23 (0.99–5.02) 0.045
Liver hepatocellular carcinoma 1.7 (1.17–2.48) 0.0052
Pancreatic ductal adenocarcinoma 1.9 (1.13–3.19) 0.014
MAP3K2 Kidney renal clear cell carcinoma 0.66 (0.49–0.89) 0.0059
Sarcoma 0.62 (0.41–0.93) 0.021
Breast cancer 1.44 (1.02–2.03) 0.037
Kidney renal papillary cell carcinoma 2.16 (1.18–3.95) 0.011
MAP3K3 Esophageal Adenocarcinoma 0.34 (0.16–0.73) 0.0036
Esophageal Squamous Cell Carcinoma 0.42 (0.18–0.97) 0.035
Head-neck squamous cell carcinoma 0.75 (0.57–0.99) 0.041
Lung adenocarcinoma 0.67 (0.5–0.91) 0.011
Pancreatic ductal adenocarcinoma 0.52 (0.34–0.78) 0.0015
Thymoma 0.29 (0.08–1.06) 0.046
Bladder carcinoma 1.39 (1.04–1.87) 0.027
Breast cancer 1.59 (1.1–2.3) 0.013
Liver hepatocellular carcinoma 1.65 (1.12–2.44) 0.01
Lung squamous cell carcinoma 1.38 (1.04–1.83) 0.024
Pheochromocytoma and Paraganglioma 5.43 (0.98–29.97) 0.03
Sarcoma 1.51 (1.01–2.26) 0.041
Testicular Germ Cell tumor 7.17 (0.74–69.24) 0.047
Uterine corpus endometrial carcinoma 2.09 (1.38–3.18) 0.00041
MAP3K4 Kidney renal clear cell carcinoma 0.64 (0.44–0.93) 0.017
Lung squamous cell carcinoma 0.69 (0.5–0.96) 0.026
Pancreatic ductal adenocarcinoma 0.61 (0.4–0.92) 0.018
Rectum adenocarcinoma 0.28 (0.1–0.84) 0.015
Breast cancer 1.64 (1.18–2.26) 0.0025
Cervical squamous cell carcinoma 1.69 (1.06 -2.71) 0.026
Esophageal Adenocarcinoma 2.11 (1.11–4) 0.02
Kidney renal papillary cell carcinoma 2.07 (1.12–3.81) 0.018
Liver hepatocellular carcinoma 1.83 (1.25–2.68) 0.0015
Ovarian cancer 1.49 (1.13–1.96) 0.0041
Stomach adenocarcinoma 1.39 (1 –1.94) 0.049
MAP3K5 Bladder carcinoma 0.66 (0.49–0.9) 0.0073
Head-neck squamous cell carcinoma 0.61 (0.46–0.82) 0.00074
Kidney renal clear cell carcinoma 0.57 (0.42–0.77) 0.00023
Lung squamous cell carcinoma 0.67 (0.47–0.94) 0.02
Rectum adenocarcinoma 0.25 (0.09–0.72) 0.0055
Sarcoma 0.59 (0.39–0.91) 0.015
Thyroid carcinoma 0.3 (0.1–0.93) 0.027
Cervical squamous cell carcinoma 1.7 (1.04–2.78) 0.032
Ovarian cancer 1.34 (1.02–1.76) 0.034
Pancreatic ductal adenocarcinoma 2.51 (1.48–4.25) 0.0004
MAP3K6 Breast cancer 0.64 (0.46–0.89) 0.0076
Cervical squamous cell carcinoma 0.49 (0.31–0.79) 0.0028
Esophageal Adenocarcinoma 0.45 (0.22–0.9) 0.02
Head-neck squamous cell carcinoma 0.63 (0.48–0.83) 0.00073
Lung adenocarcinoma 0.67 (0.48–0.93) 0.016
Stomach adenocarcinoma 0.7 (0.49 -1) 0.047
Thyroid carcinoma 0.28 (0.11–0.76) 0.0076
Uterine corpus endometrial carcinoma 0.61 (0.41–0.93) 0.02
Liver hepatocellular carcinoma 1.65 (1.14–2.39) 0.0077
Rectum adenocarcinoma 2.96 (1.29–6.79) 0.0076
Thymoma 5.54 (1.41–21.7) 0.0064
MAP3K7 Esophageal Squamous Cell Carcinoma 0.23 (0.07–0.81) 0.014
Ovarian cancer 0.76 (0.58–0.98) 0.034
Rectum adenocarcinoma 0.43 (0.19–1.01) 0.047
Thymoma 0.18 (0.05–0.85) 0.005
Cervical squamous cell carcinoma 1.81 (1.04–3.15) 0.035
Esophageal Adenocarcinoma 2.66 (1.2–5.92) 0.013
Kidney renal clear cell carcinoma 1.44 (1.07–1.94) 0.017
Kidney renal papillary cell carcinoma 1.81 (1–3.29) 0.047
Liver hepatocellular carcinoma 1.91 (1.33–2.75) 0.00035
Sarcoma 2.23 (1.47–3.38) 0.00011
MAP3K8 Bladder carcinoma 0.69 (0.51–0.93) 0.013
Breast cancer 0.68 (0.49–0.94) 0.018
Esophageal Adenocarcinoma 0.33 (0.13–0.84) 0.015
Head-neck squamous cell carcinoma 0.71 (0.54–0.92) 0.01
Lung adenocarcinoma 0.63 (0.46–0.84) 0.0019
Ovarian cancer 0.7 (0.53–0.91) 0.0075
Sarcoma 0.47 (0.31–0.7) 0.00012
Esophageal Squamous Cell Carcinoma 3.02 (1.35–6.76) 0.0049
Kidney renal clear cell carcinoma 2.15 (1.58–2.91) 0.00000048
Thymoma 6.09 (1.44–25.85) 0.0054
Thyroid carcinoma 2.73 (1.01–7.36) 0.039
MAP3K9 Kidney renal papillary cell carcinoma 0.46 (0.24–0.89) 0.019
Pancreatic ductal adenocarcinoma 0.61 (0.4–0.93) 0.021
Stomach adenocarcinoma 0.57 (0.38–0.86) 0.0068
Kidney renal clear cell carcinoma 1.41 (1.02–1.96) 0.037
Liver hepatocellular carcinoma 2.16 (1.49–3.13) 0.000032
Ovarian cancer 1.31 (1.01–1.7) 0.044
Pheochromocytoma and Paraganglioma 12.16 (1.36–108.85) 0.0043
Thymoma 4.39 (0.91–21.19) 0.044
Uterine corpus endometrial carcinoma 1.68 (1.1–2.56) 0.014
MAP3K10 Bladder carcinoma 0.56 (0.41–0.76) 0.0002
Head-neck squamous cell carcinoma 0.64 (0.49–0.84) 0.0011
Pancreatic ductal adenocarcinoma 0.51 (0.34–0.78) 0.0013
Cervical squamous cell carcinoma 1.69 (1.01–2.81) 0.043
Kidney renal clear cell carcinoma 2.07 (1.53–2.8) 0.0000017
Liver hepatocellular carcinoma 1.53 (1.03–2.26) 0.032
Thyroid carcinoma 3.77 (1.37–10.39) 0.006
Uterine corpus endometrial carcinoma 1.63 (1.07–2.46) 0.02
MAP3K11 Bladder carcinoma 0.71 (0.53–0.95) 0.02
Cervical squamous cell carcinoma 0.55 (0.35–0.88) 0.012
Kidney renal clear cell carcinoma 0.71 (0.52–0.96) 0.027
Sarcoma 0.59 (0.37–0.93) 0.022
Stomach adenocarcinoma 0.65 (0.46–0.93) 0.018
Liver hepatocellular carcinoma 1.85 (1.17–2.93) 0.0076
Lung squamous cell carcinoma 1.38 (1–1.9) 0.05
MAP3K12 Lung adenocarcinoma 0.64 (0.48–0.85) 0.0023
Pancreatic ductal adenocarcinoma 0.44 (0.29–0.68) 0.00015
Sarcoma 0.46 (0.3–0.69) 0.00014
Thymoma 0.21 (0.04–1.02) 0.034
Kidney renal clear cell carcinoma 2.33 (1.7–3.19) 0.000000062
Kidney renal papillary cell carcinoma 1.83 (1–3.34) 0.046
Pheochromocytoma and Paraganglioma 9.65 (1.12–82.77) 0.011
Stomach adenocarcinoma 1.5 (1.09–2.08) 0.013
Uterine corpus endometrial carcinoma 1.7 (1.12–2.59) 0.011
MAP3K13 Bladder carcinoma 0.69 (0.5–0.95) 0.024
Cervical squamous cell carcinoma 0.53 (0.33–0.86) 0.0084
Esophageal Adenocarcinoma 0.46 (0.23–0.92) 0.024
Kidney renal clear cell carcinoma 0.54 (0.4–0.72) 0.000032
Lung squamous cell carcinoma 0.63 (0.48–0.83) 0.00093
Ovarian cancer 0.54 (0.39–0.75) 0.00017
Rectum adenocarcinoma 0.38 (0.15–0.95) 0.031
Stomach adenocarcinoma 0.69 (0.5–0.96) 0.026
Pancreatic ductal adenocarcinoma 2.24 (1.48–3.42) 0.00011
Pheochromocytoma and Paraganglioma 5.15 (0.93–28.25) 0.036
Sarcoma 1.79 (1.12–2.86) 0.014
Thymoma 18.89 (2.33–153.14) 0.00016
Uterine corpus endometrial carcinoma 2.38 (1.56–3.62) 0.0000320
MAP3K14 Breast cancer 0.64 (0.45–0.91) 0.011
Cervical squamous cell carcinoma 0.39 (0.25–0.63) 0.000053
Head-neck squamous cell carcinoma 0.62 (0.46–0.85) 0.0026
Pancreatic ductal adenocarcinoma 0.57 (0.37–0.88) 0.011
Rectum adenocarcinoma 0.41 (0.19–0.89) 0.02
Sarcoma 0.63 (0.42–0.93) 0.019
Thyroid carcinoma 0.27 (0.09–0.79) 0.01
Kidney renal papillary cell carcinoma 2.08 (1.15–3.79) 0.014
Liver hepatocellular carcinoma 1.44 (1.02–2.03) 0.038
Thymoma 17.42 (2.17–139.68) 0.00022
MAP3K15 Pancreatic ductal adenocarcinoma 0.58 (0.39–0.89) 0.011
Esophageal Adenocarcinoma 2.95 (1.56–5.6) 0.0005
Head-neck squamous cell carcinoma 1.44 (1.08–1.92) 0.013
Kidney renal clear cell carcinoma 1.92 (1.38–2.67) 0.000088
Kidney renal papillary cell carcinoma 4.28 (2.11–8.68) 0.000011
Liver hepatocellular carcinoma 1.59 (1.12–2.27) 0.0088
Sarcoma 2.05 (1.38–3.05) 0.00031
Thyroid carcinoma 2.92 (1.02–8.43) 0.037
Uterine corpus endometrial carcinoma 3.74 (2.33–6) 0.0000000044
MAP3K16 Kidney renal clear cell carcinoma 0.62 (0.46–0.83) 0.0013
Rectum adenocarcinoma 0.39 (0.16–0.98) 0.038
Cervical squamous cell carcinoma 2.24 (1.22–4.09) 0.0072
Liver hepatocellular carcinoma 1.57 (1.11–2.22) 0.011
Stomach adenocarcinoma 1.59 (1.14–2.2) 0.0051
MAP3K17 Head-neck squamous cell carcinoma 0.66 (0.51–0.87) 0.0025
Kidney renal clear cell carcinoma 0.73 (0.54–0.98) 0.038
Kidney renal papillary cell carcinoma 0.44 (0.24–0.8) 0.006
Lung adenocarcinoma 0.63 (0.47–0.85) 0.0025
Pancreatic ductal adenocarcinoma 0.42 (0.25–0.7) 0.00057
Stomach adenocarcinoma 0.65 (0.45–0.94) 0.02
Uterine corpus endometrial carcinoma 0.46 (0.3–0.7) 0.00019
MAP3K18 Kidney renal clear cell carcinoma 0.45 (0.33–0.61) 0.00000021
Rectum adenocarcinoma 0.27 (0.08–0.89) 0.02
Sarcoma 0.61 (0.38–0.99) 0.044
Stomach adenocarcinoma 0.69 (0.49–0.96) 0.026
Thymoma 0.1 (0.01–0.85) 0.011
Kidney renal papillary cell carcinoma 2.24 (1.12–4.46) 0.019
Lung squamous cell carcinoma 1.43 (1.09–1.88) 0.0094
Pheochromocytoma and Paraganglioma 4.89 (0.89–26.99) 0.044
MAP3K19 Bladder carcinoma 0.58 (0.43–0.79) 0.00039
Breast cancer 0.67 (0.47–0.96) 0.027
Cervical squamous cell carcinoma 0.47 (0.25–0.9) 0.019
Liver hepatocellular carcinoma 0.64 (0.45–0.9) 0.011
Lung adenocarcinoma 0.68 (0.48–0.95) 0.025
Pancreatic ductal adenocarcinoma 0.6 (0.37–0.97) 0.037
Rectum adenocarcinoma 0.28 (0.08–0.95) 0.029
Uterine corpus endometrial carcinoma 0.44 (0.24–0.79) 0.0048
Kidney renal clear cell carcinoma 1.83 (1.35–2.47) 0.000064
Kidney renal papillary cell carcinoma 2.38 (1.01–5.64) 0.042
Lung squamous cell carcinoma 1.47 (1.1–1.95) 0.008
MAP3K20 Esophageal Squamous Cell Carcinoma 0.38 (0.16–0.87) 0.018
Sarcoma 0.57 (0.35–0.93) 0.022
Cervical squamous cell carcinoma 1.76 (1.08–2.87) 0.021
Kidney renal papillary cell carcinoma 2.95 (1.56–5.58) 0.00047
Lung adenocarcinoma 1.41 (1.03–1.93) 0.029
Lung squamous cell carcinoma 1.38 (1.01–1.88) 0.042
Pancreatic ductal adenocarcinoma 1.53 (1.01–2.31) 0.041
MAP3K21 Esophageal Squamous Cell Carcinoma 0.32 (0.13–0.78) 0.0086
Rectum adenocarcinoma 0.42 (0.18–0.96) 0.032
Breast cancer 1.54 (1.03–2.31) 0.034
Kidney renal papillary cell carcinoma 2.72 (1.3–5.68) 0.0056
Liver hepatocellular carcinoma 1.53 (1.07–2.17) 0.018
Uterine corpus endometrial carcinoma 1.94 (1.25–3.01) 0.0025
Significant p (<0.05) and HR values that are positively correlated with patient survival are in bold, while those italicized indicates a negative correlation. Some cancer types are not shown due to non-significant data for that gene and can be found in Supplementary Table 3.
3.2 MAP3K2

MAP3K2 is a moderately studied kinase with a PubMed score of 83.41 and a Novelty Score of –4.42 (Table 1). Overexpression of MAP3K2 has been identified in non-small cell lung cancer, hepatocellular carcinoma, prostate cancer, gastric cancer, and triple-negative breast cancer (TNBC) [19, 20, 21, 22, 23]. Previous studies demonstrated Forkhead box F1 (FOXF1), the transcriptional regulator of epithelial-mesenchymal transition (EMT), promoted tumor growth and invasion by upregulating MAP3K2 [24, 25]. Additionally, the knockdown of MAP3K2 inhibited cell migration and metastasis in several cancer types, suggesting the involvement of MAP3K2 in regulating tumor invasion and metastasis via MAP3K2-ERK5 signaling pathways [20, 21, 22]. MAP3K2 was also identified as a non-histone substrate of SET and MYND domain-containing protein 3 (SMYD3), a chromatin modifier. SMYD3-mediated lysine methylation of MAP3K2 increased the activation of MAP kinase signaling pathways and promoted Ras-driven carcinomas [26, 27]. The mRNA expression of MAP3K2 was positively associated with survival in kidney renal clear cell carcinoma and sarcoma. MAP3K2 expression was correlated with decreased survival in breast cancer and kidney renal papillary cell carcinoma (Table 2). MAP3K2 mRNA expression in other cancer types did not make the statistical cutoff.

3.3 MAP3K3

Similar to MAP3K2, MAP3K3 is a moderately studied member of the MAP3K family, with a PubMed score of 74.03 and a Novelty score of –4.13 (Table 1). MAP3K3 is involved in the development of early embryonic cardiovascular systems, endothelial cell proliferation, apoptosis, as well as inflammatory and immune responses [28, 29]. Dysregulated expression of MAP3K3 has been implicated in several cancer types, including ovarian cancer, breast cancer, kidney cancer, NSCC, and esophageal cancer [29, 30, 31, 32, 33, 34]. MAP3K3 overexpression increased activation of NF-κB signaling pathway and promoted EMT and tumor cell proliferation in ovarian cancer and breast cancer [29, 34]. Santoro et al. [31] identified MAP3K3 as a contributor to EMT and stemness in pancreatic cancer by positively regulating the oncogenic activity of yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ). This mechanism is independent of NF-κB pathway. Interestingly, He et al. [35] reported that MAP3K3 overexpression correlated with an active immune response in primary lung adenocarcinomas associated with improved patient survival [35]. In cerebral cavernous malformations, increased activity of MAP3K3 and its target genes KLF2/4 in endothelial cells were identified as causal events [28, 36]. Expression of MAP3K3 mRNA was associated with increased survival of esophageal adenocarcinoma, esophageal squamous cell carcinoma, head-neck squamous cell carcinoma, lung adenocarcinoma, pancreatic ductal adenocarcinoma, and thymoma. MAP3K3 expression was negatively associated with survival in bladder carcinoma, breast cancer, liver hepatocellular carcinoma, lung squamous cell carcinoma, pheochromocytoma and paraganglioma, sarcoma, testicular germ cell tumor, and uterine corpus endometrial carcinoma (Table 2). MAP3K3 mRNA expression in other cancer types did not make the statistical cutoff.

3.4 MAP3K4

MAP3K4 has a PubMed score of 43.58 and Novelty score of –3.82 (Table 1). Despite its relative novelty being comparable to similarly scored MAP3K16 and MAP3K17, MAP3K4 is not an IDG- eligible kinase. Previous studies reported the involvement of MAP3K4 in EMT and lactate secretion of breast cancer cells via HER2/HER3 signaling pathways [37, 38]. The role of MAP3K4 in EMT regulation via histone acetylation in trophoblast stem cells has also been demonstrated [39, 40]. MAP3K4 functions as a mediator of the stress-activated p38 MAPK pathway, whose mutation has relevance for endometrial cancer and EBV+ gastric cancer [41, 42]. MAP3K4 also contributed to proliferation and invasion of cervical cancer cells by interacting with Erb-b2 receptor tyrosine kinase 3 (ERBB3) [43]. Zhang et al. [44] identified MAP3K4 (MEKK4) to be tumor-suppressive via the MEKK4-MKK4-p38-p21signaling pathway in pancreatic cancer. While MAP3K4 was downregulated in parathyroid adenoma, it was constitutively active in urothelial carcinoma cells [45, 46]. MAP3K4 also played an important role in neuroepithelial development and loss of MAP3K4 could result in neural tube defects [47].

The mRNA expression of MAP3K4 was positively associated with survival in kidney renal clear cell carcinoma, lung squamous cell carcinoma, pancreatic ductal adenocarcinoma, and rectum adenocarcinoma. MAP3K4 expression was correlated with decreased survival in breast cancer, cervical squamous cell carcinoma, esophageal adenocarcinoma, kidney renal papillary cell carcinoma, liver hepatocellular carcinoma, ovarian cancer, stomach adenocarcinoma (Table 2). MAP3K4 mRNA expression in other cancer types did not make the statistical cutoff.

3.5 MAP3K5

MAP3K5 is the most well-studied MAP3K family member with a PubMed score of 812.85 and a Novelty score of –6.54 (Table 1). MAP3K5, also known as ASK1, is a member of the stress- induced apoptosis signal-regulating kinase (ASK) family [48]. ASK1 activates both p38 and JNK pathways responding to stressors such as cytokines, reactive oxygen species (ROS), and endoplasmic reticulum (ER) stress [49]. Thus, ASK1 plays a critical role in stress response and its dysfunction is involved in various diseases, including cancers, neurodegeneration, and cardiovascular diseases. Existing studies identified ASK1 as an antioncogene by promoting ROS- induced and ER-mediated apoptosis [49, 50]. Low expression or downregulated activity of ASK1 has been demonstrated in several cancer types, including HCC, breast cancer, and Ewing sarcoma [51, 52, 53]. However, some studies also reported that overexpression of ASK1 and its upregulated activity promotes cancer cell motility and proliferation in oral squamous cell carcinoma and ovarian cancer, and pancreatic cancer [54, 55, 56]. Therefore, ASK1 can be a therapeutic target by either activating ASK1-mediated apoptosis or inhibiting its activity based on specific cancer types. Novel triazolothiadiazines were identified as potent anticancer agents by triggering oxidative stress-induced apoptosis through ASK1 activation in HCC [57]. Selonsertib (GS-4997), an ASK1 inhibitor, attenuates multidrug resistance in cancer cells overexpressing ATP-binding cassette transporters ABCB1 and 2 [58, 59]. The mRNA expression of MAP3K5 was associated with increased survival in bladder carcinoma, head-neck squamous cell carcinoma, kidney renal clear cell carcinoma, lung squamous cell carcinoma, rectum adenocarcinoma, sarcoma, and thyroid carcinoma. MAP3K5 was expression negatively correlated with survival in cervical squamous cell carcinoma, ovarian cancer, and pancreatic ductal adenocarcinoma (Table 2). MAP3K5 mRNA expression in other cancer types did not make the statistical cutoff.

3.6 MAP3K6

MAP3K6 is one of the less studied members of the MAP3K family with a PubMed score of 27.05 and Novelty score of –5.22 (Table 1). Despite having scores similar to IDG-eligible kinases MAP3K10 and MAP3K14, MAP3K6 did not receive this designation. MAP3K6 is also a member of the ASK family, known as ASK2 [48]. Existing literature demonstrate that MAP3K6 is inhibited by CDK5 to regulate melanin production in mice [60]. Furthermore, MAP3K6 mutations are associated with cerebral small vessel disease (cSVD) causing stroke, cognitive impairment, and tremor, as well as the development of gastric cancer [61, 62]. Along with MAP3K5, MAP3K6 also known as ASK2, is a member of ASK family [49]. ASK2 has been reported to regulate tumor angiogenesis [63]. The mRNA expression of MAP3K6 was positively associated with survival in breast cancer, cervical squamous cell carcinoma, esophageal adenocarcinoma, head-neck squamous cell carcinoma, lung adenocarcinoma, stomach adenocarcinoma, thyroid carcinoma, and uterine corpus endometrial carcinoma. MAP3K6 expression was negatively associated with survival in liver hepatocellular carcinoma, rectum adenocarcinoma, and thymoma (Table 2). MAP3K6 mRNA expression in other cancer types did not make the statistical cutoff.

3.7 MAP3K7

MAP3K7 is one of the better studied members of MAP3K family with a PubMed score of 209.21 and a Novelty score of –5.22 (Table 1). MAP3K7, also known as TGF-β-activated kinase 1 (TAK1), is a critical mediator of NF-κB and JNK signaling pathways that regulate embryonic development, immune responses, and cell survival [64]. Because MAP3K7 downstream molecules NF-κB and JNK are involved in cancer cell survival and apoptosis, MAP3K7 regulates tumor initiation, proliferation, and metastasis as a cancer promoter or suppressor depending on specific receptors and cell types [65]. While specific deficiency MAP3K7 causes cell death, inflammation, fibrosis, and carcinogenesis of hepatocytes due to inhibition of NF-κB-dependent survival, higher co-expression of MAP3K7 and mTOR was positively correlated with proliferation of HCC [65, 66]. Overexpression and hyperactivation of MAP3K7 have been implicated in multiple cancers, including esophageal, thyroid, gastric, and ovarian [67, 68, 69, 70, 71]. As a critical mediator between receptors and transcription factors, MAP3K7 was identified as a potential therapeutic target for cancer therapy. Several chemical MAP3K7 inhibitors include the natural compound 5(Z)-7- oxozeaenol, LYTAK1, AZ-TAK1, Takinib, and NG25 [72, 73, 74, 75, 76].

The mRNA expression of MAP3K7 was associated with increased survival in esophageal squamous cell carcinoma, ovarian cancer, rectum adenocarcinoma, and thymoma. MAP3K7 expression was negatively associated with survival in cervical squamous cell carcinoma, esophageal adenocarcinoma, kidney renal clear cell carcinoma, kidney renal papillary cell carcinoma, liver hepatocellular carcinoma, and sarcoma (Table 2). MAP3K7 mRNA expression in other cancer types did not make the statistical cutoff.

3.8 MAP3K8

MAP3K8 is the second most-studied kinase of the MAP3K family with a PubMed score of 220.14 and a Novelty score of –5.15 (Table 1). MAP3K8, also commonly known as tumor progression locus 2 (Tpl2), regulates both innate and adaptive immunity, as well as inflammatory responses. Previous studies reported that overexpression of Tpl2 activates the ERK, JNK, and p38 MAPK pathways, as well as the transcription factors NFAT and NF-κB, and ultimately regulates the production of various cytokines [77, 78, 79, 80]. MAPK8 has variable effects on tumors and its role as both tumor suppressor and tumor promoter has been reported. For example, MAP3K8 acts as a tumor suppressor gene, and a low expression of MAP3K8 is associated with reduced lung cancer patient survival and an increase in metastasis biomarkers in skin cancer [81, 82]. However, MAP3K8 overexpression contributes to tumor proliferation, and metastasis in ovarian cancer, squamous cell carcinoma, colorectal cancer, prostate cancer, and breast cancer [83, 84, 85, 86, 87]. The mRNA expression of MAP3K8 was positively correlated with survival in bladder carcinoma, breast cancer, esophageal adenocarcinoma, head-neck squamous cell carcinoma, lung adenocarcinoma, ovarian cancer, and sarcoma. MAP3K8 expression was associated with decreased survival in esophageal squamous cell carcinoma, kidney renal clear cell carcinoma, thymoma, and thyroid carcinoma (Table 2). MAP3K8 mRNA expression in other cancer types did not make the statistical cutoff.

3.9 MAP3K9

MAP3K9 has a PubMed score of 43.94 and a Novelty score of –3.72 (Table 1). Despite its relative novelty being comparable to similarly scored MAP3K16 and MAP3K17, MAP3K9 is not an IDG- eligible kinase. MAP3K9, also known as mixed lineage kinase I (MLK1), belongs to the MLK family, which are upstream activators of MEK/ERK and JNK pathways [88]. While the role of MAP3K9 in cancer is not well-defined, previous studies reported that targeting MAP3K9 using microRNA suppressed tumor progression in pancreatic cancer, pharyngolaryngeal cancer, HCC, and esophagus squamous cell carcinoma, suggesting its involvement in cancer pathogenesis [89, 90, 91]. In lung cancer cells, gain-of-function mutation MAP3K9 leads to the increased activation of downstream ERK pathway, which potentially promotes tumor proliferation [92]. Marusiak et al. [93] demonstrated MAP3K9 (MLK1) reactivates MEK/ERK pathway independently of RAF, contributing to the resistance of RAF inhibitors in melanoma. Additionally, MAP3K9 has been identified as a gene that is frequently mutated in metastatic melanoma [94]. The mRNA expression of MAP3K9 was positively correlated with survival in kidney renal papillary cell carcinoma, pancreatic ductal adenocarcinoma, and stomach adenocarcinoma. MAP3K9 expression was negatively correlated with survival in kidney renal clear cell carcinoma, liver hepatocellular carcinoma, ovarian cancer, pheochromocytoma and paraganglioma, thymoma, and uterine corpus endometrial carcinoma (Table 2). MAP3K9 mRNA expression in other cancer types did not make the statistical cutoff.

3.10 MAP3K10

MAP3K10 is an IDG-eligible kinase with a PubMed score of 25.78 and Novelty score of –3.34 (Table 1). MAP3K is also a member of the MLK family, known as MLK2 [88]. Existing literature identifies MAP3K10 as a mediator of TGFß activation with a role in regulating atherosclerotic inflammatory responses [95, 96]. Furthermore, MAP3K10 has been implicated to play a role in pancreatic cancer, esophageal carcinoma, and osteosarcoma [97]. Additionally, targeting MAP3K10 with microRNA MiR-146b-3p and MiR-155-5p has been demonstrated to, respectively, abrogate pancreatic cancer stem-cell proliferation and sensitize esophageal carcinoma cells to radiation and chemotherapy [98, 99]. The mRNA expression of MAP3K10 was associated with increased survival in bladder carcinoma, head-neck squamous cell carcinoma, and pancreatic ductal adenocarcinoma. MAP3K10 expression was negatively associated with survival in cervical squamous cell carcinoma, kidney renal clear cell carcinoma, liver hepatocellular carcinoma, thyroid carcinoma, and uterine corpus endometrial carcinoma (Table 2). MAP3K10 mRNA expression in other cancer types did not make the statistical cutoff.

3.11 MAP3K11

MAP3K11 is a moderately studied member of the MAP3K family with a PubMed score of 117.01 and a Novelty score of –4.5 (Table 1). MAP3K1, also known as MLK3, has been shown to activate p38 pathway besides MEK/ERK and JNK pathways [88]. The role of MLK3 (MAP3K11) in cancer cell migration has been demonstrated in several cancer types, including breast and lung cancers [100, 101, 102]. Chen et al. [102] reported a crucial role of MLK3 (MAP3K11) in cell migration in breast cancer by activating JNK signaling to AP-1, which promotes EMT and an invasive breast cancer phenotype [102]. Another proposed mechanism of MLK3 (MAP3K11) regulating cancer cell migration is through dysregulating mediators critical for cytoskeletal rearrangement and focal adhesion dynamics, including Cdc42, Rac1, and RhoA GTPases [100, 101, 103]. In prostate cancer MLK3 (MAP3K11) also facilitates the collagen type I-induced EMT switch, leading to JNK- mediated increased expression of N-cadherin, an EMT marker associated with promoting migratory and invasive capacity [103, 104]. Ma et al. [105] showed that MLK3 expression is upregulated in cervical cancer cells and MLK3 blocking suppresses cancer progression via autophagy- dependent apoptosis. Additionally, targeting MAP3K11 using microRNA inhibited tumor proliferation in NSCLC and esophageal cancer [106, 107]. Similar to MAP3K9 (MLK1), MAP3K11 (MLK3) also promotes resistance to RAF inhibitor vemurafenib by reactivating MEK/ERK pathway independently of RAF, contributing to cell survival and progression in melanoma [106]. MLK3 also plays a role in inflammation by regulating NF-κB/NLRP3 signaling pathway-mediated inflammation and JNK/p53 signaling pathway-mediated oxidative stress, which are associated with myocardial fibrosis [108]. The mRNA expression of MAP3K11 was correlated with increased survival in bladder carcinoma, cervical squamous cell carcinoma, kidney renal clear cell carcinoma, sarcoma, and stomach adenocarcinoma. MAP3K11 expression was associated with decreased survival in liver hepatocellular carcinoma, lung squamous cell carcinoma (Table 2). MAP3K11 mRNA expression in other cancer types did not make the statistical cutoff.

3.12 MAP3K12

MAP3K12 is a moderately studied member of the MAP3K family with a PubMed score of 157.61 and a Novelty score of –5.07 (Table 1). MAP3K12, also known as dual leucine zipper kinase (DLK), is a member of MLK family [88]. It has been investigated in the pathogenesis of neurodegenerative diseases and diabetes mellitus, its role in cancer has not been as well-studied in cancer. Yu et al. [109] demonstrated that targeting MAP3K12 using microRNA miR-150-5p suppresses cell proliferation and invasion in prostate cancer cells. MAP3K12 is also upregulated in T-cell acute lymphoblastic leukemia (T-ALL), suggesting that MAP3K12 could be a marker of T-ALL for future studies [110]. DLK (MAP3K12) regulates the stress-induced JNK signaling in neurons and has been identified as a central regulation of various neuronal degradation models [111, 112, 113, 114, 115, 116]. In diabetes mellitus, MAP3K12-mediated JNK signaling pathway can underlie endothelial dysfunction, suggesting MAP3K12 as a potential therapeutic target [117]. The mRNA expression of MAP3K12 was positively correlated with lung adenocarcinoma, pancreatic ductal adenocarcinoma, sarcoma, and thymoma. MAP3K12 expression was negatively associated with survival in kidney renal clear cell carcinoma, kidney renal papillary cell carcinoma, pheochromocytoma and paraganglioma, stomach adenocarcinoma, and uterine corpus endometrial carcinoma (Table 2). MAP3K12 mRNA expression in other cancer types did not make the statistical cutoff.

3.13 MAP3K13

MAP3K13 is one of the better-studied members of the MAP3K family with a PubMed score of 200.04 and a Novelty score of –5.38 (Table 1). MAP3K13, also known as leucine zipper-bearing kinase (LZK), belongs to the MLK family and has a high sequence identity to DLK/MAPK312 [88]. LZK was shown to regulate NF-κB and JNK signaling pathways, which could be cancer promoting [118, 119]. In breast cancer, MAP3K13 overexpression stabilizes and enhances the transcriptional activity of Myc oncogene, contributing to poor patient survival [120]. Amplified MAP3K13 promotes cancer cell viability and proliferation by maintaining expression of gain-of- function mutant p53 [121]. Additionally, Fu et al. [122] reported that long non-coding RNAs (lncRNA) LINC01287 activated NF-κB signaling through regulating MAP3K13, potentially regulating migration, invasion, and EMT in colon cancer [122]. More recently, LZK has been identified as a novel positive regulator of axon growth [123]. The mRNA expression of MAP3K13 was positively associated with survival in bladder carcinoma, cervical squamous cell carcinoma, esophageal adenocarcinoma, kidney renal clear cell carcinoma, lung squamous cell carcinoma, ovarian cancer, rectum adenocarcinoma, and stomach adenocarcinoma. MAP3K13 expression was associated with decreased survival in pancreatic ductal adenocarcinoma, pheochromocytoma, and paraganglioma, sarcoma, thymoma, and uterine corpus endometrial carcinoma (Table 2). MAP3K13 mRNA expression in other cancer types did not make the statistical cutoff.

3.14 MAP3K14

MAP3K14 is an IDG-eligible kinase with a PubMed score of 29.23 and Novelty score of –3.35 (Table 1). It has been described, in mantle cell lymphoma (MCL), as a mediator in the non-canonical NfκB pathway. Mutations in the NFκB pathway leads to dependence of MAP3K14, both in vitro and in vivo, suggesting that MAP3K14 is potentially a therapeutic target in MCL [124]. Additionally, MAP3K14 has been identified to be a regulator of the innate and adaptive immune responses with mutations leading to atypical-combined immunodeficiency [125, 126]. The mRNA expression of MAP3K14 was associated with increased survival in breast cancer, cervical squamous cell carcinoma, head-neck squamous cell carcinoma, pancreatic ductal adenocarcinoma, rectum adenocarcinoma, sarcoma, and thyroid carcinoma. MAP3K14 expression was negatively associated with survival in kidney renal papillary cell carcinoma, liver hepatocellular carcinoma, and thymoma (Table 2). MAP3K14 mRNA expression in other cancer types did not make the statistical cutoff.

3.15 MAP3K15

MAP3K15 is an IDG-eligible kinase with a PubMed score of 10.11 and Novelty score of –2.11 (Table 1). MAP3K15 is also known as ASK3, a member of the ASK family [49]. It has been identified to play an important role in immune-related activities against cancer with one study demonstrating a correlation between MAP3K15 expression and immune infiltration in osteosarcoma [127]. Despite this report, other studies found that high levels of MAP3K15 to be correlated with poor prognosis in Osteosarcoma and uterine cancer [127, 128]. More broadly, studies on MAP3K15 have shown that it has protective effects against osmotically driven hypertension, and a knockdown phenotype of MAP3K19 results in an inherited form of hypertension [129].

The mRNA expression of MAP3K15 was positively associated with pancreatic ductal adenocarcinoma. MAP3K15 expression was associated with decreased survival in esophageal adenocarcinoma, head-neck squamous cell carcinoma, kidney renal clear cell carcinoma, kidney renal papillary cell carcinoma, liver hepatocellular carcinoma, sarcoma, thyroid carcinoma, and uterine corpus endometrial carcinoma (Table 2). MAP3K15 mRNA expression in other cancer types did not make the statistical cutoff.

3.16 MAP3K16 (TAOK1)

Better known as TAOK1, MAP3K16 is the most characterized IDG-eligible MAP3K member with a PubMed score of 57.97 and Novelty score of –4.07 (Table 1). Existing literature identifies TAOK1 involvement in neurodevelopment with dysregulation and de-novo variants leading to neurodevelopmental disorders [130, 131]. Furthermore, MAP3K16 is a positive regulator of TLR4-induced inflammatory responses, activating macrophages through promotion of ERK1/2 [132]. The mRNA expression of TAOK1 was associated with increased survival in kidney renal clear cell carcinoma and rectum adenocarcinoma. TAOK1 expression was negatively associated with survival in cervical squamous cell carcinoma, liver hepatocellular carcinoma, and stomach adenocarcinoma (Table 2). TAOK1 mRNA expression in other cancer types did not make the statistical cutoff.

3.17 MAP3K17 (TAOK2)

Better known as TAOK2, MAP3K17 is one of the better characterized IDG-eligible MAP3K members, second to MAP3K16, with a PubMed score of 46.59 and Novelty score of –3.76 (Table 1). Existing literature identifies TAOK2 as an ER-localized kinase that acts to catalyze ER-microtubule interactions [133]. Furthermore, TAOK2 has been demonstrated to play a role in neurodevelopment and cognition with studies showing control over behavioral responses to ethanol, in mice, and the development of autism spectrum disorder through RhoA signaling [134, 135]. The mRNA expression of TAOK2 was positively associated with survival in head-neck squamous cell carcinoma, kidney renal clear cell carcinoma, kidney renal papillary cell carcinoma, lung adenocarcinoma, pancreatic ductal adenocarcinoma, stomach adenocarcinoma, and uterine corpus endometrial carcinoma. TAOK2 expression did not correlate with decreased survival in any studied cancer types (Table 2). TAOK2 mRNA expression in other cancer types did not make the statistical cutoff.

3.18 MAP3K18 (TAOK3)

Better known as TAOK3, MAP3K18 has a PubMed score of 50.8 and a Novelty score of –3.88 (Table 1). Despite its relative novelty being comparable to similarly scored MAP3K16 and MAP3K17, TAOK3 is not an IDG-eligible kinase. Existing literature identifies TAOK3 as a contributing regulator of osteoblast differentiation and skeletal mineralization, lipid partitioning in the liver, and t-cell receptor signaling [136, 137, 138, 139]. Furthermore, TAOK3 has also been identified to regulate cancer stem-cells in pancreatic cancer and enhance microtubule-targeted drug resistance in breast cancer through NF-κB signaling [140, 141]. The mRNA expression of TAOK3 was positively correlated with survival in kidney renal clear cell carcinoma, rectum adenocarcinoma, sarcoma, stomach adenocarcinoma, and thymoma. TAOK3 was negatively correlated with survival in kidney renal papillary cell carcinoma, lung squamous cell carcinoma, and pheochromocytoma and paraganglioma (Table 2). TAOK3 mRNA expression in other cancer types did not make the statistical cutoff.

3.19 MAP3K19

MAP3K19 is the least studied MAP3K family member with a PubMed Score of 4.83 and Novelty Score of –1.31 (Table 1). Despite having the lowest number of publications available and the highest novelty, MAP3K19 was not among the list of IDG-eligible MAP3K members. Existing literature identifies a role for MAP3K19 in lung pathology. Boehme et al. [142, 143] identified MAP3K19 as a novel TGFß regulator in pulmonary fibrosis and as a central mediator of cigarette smoke induced pulmonary inflammation. Furthermore, Hoang et al. [144] and Jones et al. [145] identified MAP3K19 as a mediator for idiopathic pulmonary fibrosis and KRAS-mutant lung cancer. The mRNA expression of MAP3K19 was positively associated with survival in bladder carcinoma, breast cancer, cervical squamous cell carcinoma, liver hepatocellular carcinoma, lung adenocarcinoma, pancreatic ductal adenocarcinoma, rectum adenocarcinoma, and uterine corpus endometrial carcinoma. MAP3K19 expression was associated with decreased survival in kidney renal clear cell carcinoma, kidney renal papillary cell carcinoma, and lung squamous cell carcinoma (Table 2). MAP3K19 mRNA expression in other cancer types did not make the statistical cutoff.

3.10 MAP3K20

MAP3K20 is among the least characterized MAP3K family of kinases with a PubMed score of 16.1 and Novelty score of –2.79 (Table 1). Despite its relative novelty being comparable to similarly scored MAP3K10 and MAP3K21, MAP3K20 is not an IDG-eligible kinase. Existing literature identifies MAP3K20 as an ERK and JNK activator with multiple isoforms. Some isoforms of MAP3K20 have been identified to be positively correlated with gastric and colorectal cancer development while others have been demonstrated to have anti-tumor roles by promoting apoptosis in osteosarcoma [146, 147, 148, 149]. The mRNA expression of MAP3K20 was associated with increased survival in esophageal squamous cell carcinoma and sarcoma. MAP3K20 expression was negatively associated with survival in cervical squamous cell carcinoma, kidney renal papillary cell carcinoma, lung adenocarcinoma, lung squamous cell carcinoma, and pancreatic ductal adenocarcinoma (Table 2). MAP3K20 mRNA expression in other cancer types did not make the statistical cutoff.

3.21 MAP3K21

MAP3K21 is one of the least studied MAP3K family members, second to MAP3K19, with a PubMed Score of 11.8 and Novelty Score of –1.96 (Table 1). Additionally, MAP3K21 is an NIH designated IDG-eligible kinase. Existing reports identify a correlation between MAP3K21 and pediatric obesity, E. coli induced diarrhea, and disease resistance in African chickens [150, 151, 152].

Additionally, MAP3K21 has been found to be cancer promoting in breast cancer, hepatocellular carcinoma, colorectal carcinoma, ovarian cancer, and gliomas [153, 154, 155, 156, 157, 158, 159, 160]. The mRNA expression of MAP3K21 was positively associated with survival in esophageal squamous cell carcinoma and rectum adenocarcinoma. MAP3K21 expression was negatively associated with survival in breast cancer, kidney renal papillary cell carcinoma, liver hepatocellular carcinoma, and uterine corpus endometrial carcinoma (Table 2). MAP3K21 mRNA expression in other cancer types did not make the statistical cutoff.

4. Discussion

While performing a chemical compound based kinase screen, we identified MAP3K19 as a lead target in our cell models and have published a review comparing MAP3K19 expression levels in different normal and cancerous tissue types [161]. We then decided to take a broader look at the MAPK family and the lesser known MAP3K members. Mitogen-activated protein kinase (MAPK) pathways are signaling networks that regulate crucial cellular processes such as growth, proliferation, differentiation, migration, and apoptosis. MAP3Ks are upstream MAPK serine/threonine kinases that directly activate MAP2Ks through protein phosphorylation, leading to the activation of downstream MAPKs and their associated effects. This review evaluates the MAP3K family members (1 through 21) using PubMed and Novelty scores, current literature, and a critical analysis of isoform expression level with patient survival outcomes across multiple cancer types. Furthermore, this review develops a comparative characterization for each MAP3K member and provides the scientific community with a bioinformatic basis for further investigation of MAP3K signaling.

Kinase inhibitors are a major advancement in clinical therapies and have transformed disease management. Despite enormous successes with kinase inhibitors and dedicated research effort into kinase signaling, the majority of the kinome remains understudied [162, 163]. To address this gap, the NIH launched the “Illuminating the Druggable Genome” (IDG) Program in 2014. This program’s purpose is to fund pilot projects to generate additional data and tools around understudied proteins. Furthermore, this program led to the development of Pharos, an informatics database that integrates information from various sources to generate PubMed and Novelty scores for each kinase. In this review, we reference RFA-RM-21-012 to identify the list of IDG-eligible MAP3K members. Among the 21 MAP3K members, only 6—MAP3K10, MAP3K14, MAP3K15, MAP3K16, MAP3K17, and MAP3K21—members were IDG-eligible. The PubMed and Novelty scores of the IDG-eligible MAP3K members range from 11.8 to 57.97 and –4.07 to –1.96, respectively. Interestingly, MAP3K4, MAP3K6, MAP3K9, MAP3K18, MAP3K19, and MAP3K20 were not eligible despite having PubMed scores well below the highest scored IDG-eligible MAP3K16 (57.97), indicating less available publications for these 6 kinases contrasted to MAP3K16. An analogous correlation is seen when comparing Novelty scores for the same 6 kinases; higher novelty scores are seen when compared to the lowest scored MAP3K: MAP3K16 (–4.07). This relationship is especially surprising for MAP3K19, a MAP3K member with the lowest PubMed score (4.83) and highest Novelty score (–1.31) (Table 1). Altogether, this leads us to anticipate future funding opportunity announcements including MAP3K4, MAP3K6, MAP3K9, MAP3K18, MAP3K19, and MAP3K20 in the list of IDG-eligible kinases.

The MAPK pathway is a signaling pathway that is a crucial regulator of a diverse multitude of cellular processes. Because of the ubiquitous nature of MAPK signaling in cellular biology, we decided to analyze the correlations between mRNA expression levels of different MAP3K members and patient survival of different cancer types. Using the KMPlotter database, we generated a table of different MAP3K’s and their corresponding hazard ratios for different cancers. Overall, the major conclusion is that MAP3K’s play different roles in different cancers. For example: MAP3K1 expression seems to have a pro-tumor role in pancreatic ductal adenocarcinoma, MAP3K3 demonstrates antitumor effects, and MAP3K2 demonstrates no correlation. This phenomenon provides supporting evidence for the broad biological roles of MAP3K signaling. Another example is that MAP3K1 has a beneficial hazard ratio in squamous cell carcinomas of the cervix, esophagus, and of head-neck origins while having a harmful hazard ratio in kidney renal papillary cell and liver hepatocellular carcinomas. This provides supporting evidence the tissue dependency of MAP3K functions with squamous cell tumors having the most benefit from MAP3K1 expression.

One potential drawback to this bioinformatic analysis approach is that correlations were generated from tissue mRNA levels. Because mRNA expression does not always perfectly reflect protein expression levels or post-translational activity, KMPlotter correlations may not perfectly align with existing literature or pathophysiology. For example, MAP3K8 mRNA overexpression is correlated with improved ovarian cancer patient survival rates despite existing literature suggesting a pro- tumor role [87]. This quandary can be addressed by analyzing protein expression databases for supporting data. However, that process also has limitations because protein expression does not always directly correlate with protein activity. So regardless of bioinformatics technique used, scientific rigor must be addressed with supplemental validation and functional studies.

5. Conclusions

To summarize, our intent with this manuscript is to highlight the importance of the MAPK signaling pathway with an emphasis on the MAP3K family of kinases. We do this by discussing the most recent literature for each kinase, emphasizing its roles in pathology and associated signaling mechanisms. Additionally, we also use the KMPlotter bioinformatics database to generate correlations between mRNA expression levels and patient survival in different cancer types. Lastly, we discuss the PubMed and Novelty scores for each kinase and compare them to the previous IDG-eligible kinases. This comparison leads us to predict MAP3K4, MAP3K6, MAP3K9, MAP3K18, MAP3K19, and MAP3K20 to receive IDG-eligible designation in the future due to their relatively low PubMed and high Novelty scores. Overall, the MAPK pathway is diverse, complex, and ubiquitous in essential cellular processes. Disruption of this signaling axis can lead to diseases such as cancers. This review is focused on the less studied MAP3K family of the MAPK signaling pathway. Our goal is to establish a foundation for future MAP3K research by providing a summary of existing literature, preliminary bioinformatics data, and discussing potential funding sources through the NIH Illuminating the Druggable Genome program.

Author Contributions

This manuscript idea was originally conceived by KN and MEB and refined with the help of DHD, SBL, and BMC-B. KN and MNT did the majority of the research, database analysis, and writing. AR, TC, GOW, AC, JEC, and PTF contributed to the writing, data analysis and interpretation, and data presentation.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

Thank you to the Tulane DeBakey Scholars Program for providing Khoa Nguyen, and other medical students, the opportunity to pursue a longitudinal summer research experience.

Funding

Research reported in this publication was supported by the National Center for Advancing Translational Sciences of the National Institutes of Health under award number TL1R003106. This content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Chen Z, Gibson TB, Robinson F, Silvestro L, Pearson G, Xu B, et al. MAP kinases. Chemical Reviews. 2001; 101: 2449–2476.
[2]
Cuarental L, Sucunza-Sáenz D, Valiño-Rivas L Fernandez-Fernandez B, Sanz AB, Ortiz A, et al. MAP3K kinases and kidney injury. Nefrología. 2019; 39: 568–580.
[3]
Cuevas BD, Abell AN, Johnson GL. Role of mitogen-activated protein kinase kinase kinases in signal integration. Oncogene. 2007; 26: 3159–3171.
[4]
Prahallad A, Sun C, Huang S, Di Nicolantonio F, Salazar R, Zecchin D, et al. Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature. 2012; 483: 100–103.
[5]
Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, et al. Mutations of the BRAF gene in human cancer. Nature. 2002; 417: 949–954.
[6]
Di Nicolantonio F, Martini M, Molinari F, Sartore-Bianchi A, Arena S, Saletti P, et al. Wild-type BRAF is required for response to panitumumab or cetuximab in metastatic colorectal cancer. Journal of Clinical Oncology. 2008; 26: 5705–5712.
[7]
Nikiforova MN, Kimura ET, Gandhi M, Biddinger PW, Knauf JA, Basolo F, et al. BRAF mutations in thyroid tumors are restricted to papillary carcinomas and anaplastic or poorly differentiated carcinomas arising from papillary carcinomas. Journal of Clinical Endocrinology and Metabolism. 2003; 88: 5399–5404.
[8]
Braicu C, Buse M, Busuioc C, Drula R, Gulei D, Raduly L, et al. A Comprehensive Review on MAPK: A Promising Therapeutic Target in Cancer. Cancers. 2019; 11: 1618.
[9]
Nagy Á, Munkácsy G, Győrffy B. Pancancer survival analysis of cancer hallmark genes. Scientific Reports. 2021; 11: 6047.
[10]
Lánczky A, Győrffy B. Web-Based Survival Analysis Tool Tailored for Medical Research (KMplot): Development and Implementation. Journal of Medical Internet Research. 2021; 23: e27633.
[11]
Suddason T, Gallagher E. A RING to rule them all? Insights into the Map3k1 PHD motif provide a new mechanistic understanding into the diverse roles of Map3k1. Cell Death and Differentiation. 2015; 22: 540–548.
[12]
Pham TT, Angus SP, Johnson GL. MAP3K1: Genomic Alterations in Cancer and Function in Promoting Cell Survival or Apoptosis. Genes and Cancer. 2013; 4: 419–426.
[13]
Schlesinger TK, Bonvin C, Jarpe MB, Fanger GR, Cardinaux JR, Johnson GL, et al. Apoptosis stimulated by the 91-kDa caspase cleavage MEKK1 fragment requires translocation to soluble cellular compartments. Journal of Biological Chemistry. 2002; 277: 10283–10291.
[14]
Xue Z, Vis DJ, Bruna A, Sustic T, van Wageningen S, Batra AS, et al. MAP3K1 and MAP2K4 mutations are associated with sensitivity to MEK inhibitors in multiple cancer models. Cell Research. 2018; 28: 719–729.
[15]
Avivar-Valderas A, McEwen R, Taheri-Ghahfarokhi A, Carnevalli LS, Hardaker EL, Maresca M, et al. Functional significance of co-occurring mutations in PIK3CA and MAP3K1 in breast cancer. Oncotarget. 2018; 9: 21444–21458.
[16]
Koboldt DC, Fulton RS, McLellan MD, Schmidt H, Kalicki-Veizer J, McMichael JF, et al. Comprehensive molecular portraits of human breast tumours. Nature. 2012; 490: 61–70.
[17]
Wang J, Zuo J, Wahafu A, Wang M, Li R, Xie W. Combined elevation of TRIB2 and MAP3K1 indicates poor prognosis and chemoresistance to temozolomide in glioblastoma. CNS Neuroscience and Therapeutics. 2020; 26: 297–308.
[18]
Zhou W, Huang S, Jiang Q, Yuan T. Suppression of miR-4735-3p in androgen receptor-expressing prostate cancer cells increases cell death during chemotherapy. American Journal of Translational Research. 2017; 9: 3714–3722.
[19]
Wu J, Li WZ, Huang ML, Wei HL, Wang T, Fan J, et al. Regulation of cancerous progression and epithelial-mesenchymal transition by miR-34c-3p via modulation of MAP3K2 signaling in triple-negative breast cancer cells. Biochemical and Biophysical Research Communications. 2017; 483: 10–16.
[20]
Huang T, She K, Peng G, Wang W, Huang J, Li J, et al. MicroRNA-186 suppresses cell proliferation and metastasis through targeting MAP3K2 in non-small cell lung cancer. International Journal of Oncology. 2016; 49: 1437–1444.
[21]
Zhang W, Kong G, Zhang J, Wang T, Ye L, Zhang X. MicroRNA-520b inhibits growth of hepatoma cells by targeting MEKK2 and cyclin D1. PLoS ONE. 2012; 7: e31450.
[22]
Zhang Y, Wang D, Zhu T, Yu J, Wu X, Lin W, et al. CircPUM1 promotes hepatocellular carcinoma progression through the miR-1208/MAP3K2 axis. Journal of Cellular and Molecular Medicine. 2021; 25: 600–612.
[23]
Liao X, Wen L, Luo L. The Effect and Mechanism of lncRNA NR2F1-as1/miR-493-5p/MAP3K2 Axis in the Progression of Gastric Cancer. Journal of Oncology. 2021; 2021: 3881932.
[24]
Fulford L, Milewski D, Ustiyan V, Ravishankar N, Cai Y, Le T, et al. The transcription factor FOXF1 promotes prostate cancer by stimulating the mitogen-activated protein kinase ERK5. Science Signaling. 2016; 9: ra48.
[25]
Balli D, Ustiyan V, Zhang Y, Wang IC, Masino AJ, Ren X, et al. Foxm1 transcription factor is required for lung fibrosis and epithelial-to-mesenchymal transition. EMBO Journal. 2013; 32: 231–244.
[26]
Mazur PK, Reynoird N, Khatri P, Jansen PW, Wilkinson AW, Liu S, et al. SMYD3 links lysine methylation of MAP3K2 to Ras-driven cancer. Nature. 2014; 510: 283–287.
[27]
Bernard BJ, Nigam N, Burkitt K, Saloura V. SMYD3: a regulator of epigenetic and signaling pathways in cancer. Clinical Epigenetics. 2021; 13: 45.
[28]
Zhou Z, Rawnsley DR, Goddard LM, Pan W, Cao XJ, Jakus Z, et al. The cerebral cavernous malformation pathway controls cardiac development via regulation of endocardial MEKK3 signaling and KLF expression. Developmental Cell. 2015; 32: 168–180.
[29]
Zhang Y, Wang SS, Tao L, Pang LJ, Zou H, Liang WH, et al. Overexpression of MAP3K3 promotes tumour growth through activation of the NF-κB signalling pathway in ovarian carcinoma. Scientific Reports. 2019; 9: 8401.
[30]
Zhao L, Ni X, Zhao L, Zhang Y, Jin D, Yin W, et al. MiroRNA-188 Acts as Tumor Suppressor in Non-Small-Cell Lung Cancer by Targeting MAP3K3. Molecular Pharmaceutics. 2018; 15: 1682–1689.
[31]
Santoro R, Zanotto M, Carbone C, Piro G, Tortora G, Melisi D. MEKK3 Sustains EMT and Stemness in Pancreatic Cancer by Regulating YAP and TAZ Transcriptional Activity. Anticancer Research. 2018; 38: 1937–1946.
[32]
Hasan R, Sharma R, Saraya A, Chattopadhyay TK, DattaGupta S, Walfish PG, et al. Mitogen activated protein kinase kinase kinase 3 (MAP3K3/MEKK3) overexpression is an early event in esophageal tumorigenesis and is a predictor of poor disease prognosis. BMC Cancer. 2014; 14: 2.
[33]
Jia W, Dong Y, Tao L, Pang L, Ren Y, Liang W, et al. MAP3K3 overexpression is associated with poor survival in ovarian carcinoma. Human Pathology. 2016; 50: 162–169.
[34]
Fan Y, Ge N, Wang X, Sun W, Mao R, Bu W, et al. Amplification and over-expression of MAP3K3 gene in human breast cancer promotes formation and survival of breast cancer cells. Journal of Pathology. 2014; 232: 75–86.
[35]
He Y, Wang L, Liu W, Zhong J, Bai S, Wang Z, et al. MAP3K3 expression in tumor cells and tumor-infiltrating lymphocytes is correlated with favorable patient survival in lung cancer. Scientific Reports. 2015; 5: 11471.
[36]
Zhou Z, Tang AT, Wong WY, Bamezai S, Goddard LM, Shenkar R, et al. Cerebral cavernous malformations arise from endothelial gain of MEKK3–KLF2/4 signalling. Nature. 2016; 532: 122–126.
[37]
Garcia-Flores AE, Sollome JJ, Thavathiru E, Bower JL, Vaillancourt RR. HER2/HER3 regulates lactate secretion and expression of lactate receptor mRNA through the MAP3K4 associated protein GIT1. Scientific Reports. 2019; 9: 10823.
[38]
Sollome JJ, Thavathiru E, Camenisch TD, Vaillancourt RR. Her2/her3 regulates extracellular acidification and cell migration through MTK1 (MEKK4). Cellular Signalling. 2014; 26: 70–82.
[39]
Mobley RJ, Raghu D, Duke LD, Abell-Hart K, Zawistowski JS, Lutz K, et al. MAP3K4 Controls the Chromatin Modifier HDAC6 during Trophoblast Stem Cell Epithelial-to-Mesenchymal Transition. Cell Reports. 2017; 18: 2387–2400.
[40]
Abell AN, Jordan NV, Huang W, Prat A, Midland AA, Johnson NL, et al. MAP3K4/CBP-regulated H2B acetylation controls epithelial-mesenchymal transition in trophoblast stem cells. Cell Stem Cell. 2011; 8: 525–537.
[41]
Liang Q, Yao X, Tang S, Zhang J, Yau TO, Li X, et al. Integrative identification of Epstein-Barr virus-associated mutations and epigenetic alterations in gastric cancer. Gastroenterology. 2014; 147: 1350–1362.e4.
[42]
Yang L, Gao Q, Shi J, Wang Z, Zhang Y, Gao P, et al. Mitogen-activated protein kinase kinase kinase 4 deficiency in intrahepatic cholangiocarcinoma leads to invasive growth and epithelial-mesenchymal transition. Hepatology. 2015; 62: 1804–1816.
[43]
Du J, Zhou S, Wang L, Yu M, Mei L. Downregulation of ERBB3 decreases the proliferation, migration and invasion of cervical cancer cells though the interaction with MTK-1. Oncology Letters. 2018; 16: 3453–3458.
[44]
Zhang B, Meng M, Xiang S, Cao Z, Xu X, Zhao Z, et al. Selective activation of tumor-suppressive MAPKP signaling pathway by triptonide effectively inhibits pancreatic cancer cell tumorigenicity and tumor growth. Biochemical Pharmacology. 2019; 166: 70–81.
[45]
Swiatkowski S, Seifert H, Steinhoff C, Prior A, Thievessen I, Schliess F, et al. Activities of MAP-kinase pathways in normal uroepithelial cells and urothelial carcinoma cell lines. Experimental Cell Research. 2003; 282: 48–57.
[46]
Arya AK, Singh P, Saikia UN, Sachdeva N, Dahiya D, Behera A, et al. Dysregulated mitogen-activated protein kinase pathway mediated cell cycle disruption in sporadic parathyroid tumors. Journal of Endocrinological Investigation. 2020; 43: 247–253.
[47]
Chi H, Sarkisian MR, Rakic P, Flavell RA. Loss of mitogen-activated protein kinase kinase kinase 4 (MEKK4) results in enhanced apoptosis and defective neural tube development. Proceedings of the National Academy of Sciences of the United States of America. 2005; 102: 3846–3851.
[48]
Takeda K, Noguchi T, Naguro I, Ichijo H. Apoptosis Signal-Regulating Kinase 1 in Stress and Immune Response. Annual Review of Pharmacology and Toxicology. 2008; 48: 199–225.
[49]
Ryuno H, Naguro I, Kamiyama M. ASK family and cancer. Advances in Biological Regulation. 2017; 66: 72–84.
[50]
Madan E, Gogna R, Kuppusamy P, Bhatt M, Mahdi AA, Pati U. SCO2 induces p53-mediated apoptosis by Thr845 phosphorylation of ASK-1 and dissociation of the ASK-1-Trx complex. Molecular and Cellular Biology. 2013; 33: 1285–1302.
[51]
Jiang CF, Wen LZ, Yin C, Xu WP, Shi B, Zhang X, et al. Apoptosis signal-regulating kinase 1 mediates the inhibitory effect of hepatocyte nuclear factor-4α on hepatocellular carcinoma. Oncotarget. 2016; 7: 27408–27421.
[52]
Won M, Park KA, Byun HS, Sohn KC, Kim YR, Jeon J, et al. Novel anti-apoptotic mechanism of a20 through targeting ASK1 to suppress TNF-induced JNK activation. Cell Death and Differentiation. 2010; 17: 1830–1841.
[53]
Zhuo R, Kosak KM, Sankar S, Wiles ET, Sun Y, Zhang J, et al. Targeting Glutathione S-transferase M4 in Ewing sarcoma. Frontiers in Pediatrics. 2014; 2: 83.
[54]
Berschneider B, Königshoff M. WNT1 inducible signaling pathway protein 1 (WISP1): a novel mediator linking development and disease. International Journal of Biochemistry and Cell Biology. 2011; 43: 306–309.
[55]
Luo Y, Gao S, Hao Z, Yang Y, Xie S, Li D, et al. Apoptosis signal-regulating kinase 1 exhibits oncogenic activity in pancreatic cancer. Oncotarget. 2016; 7: 75155–75164.
[56]
Yin M, Zhou HJ, Zhang J, Lin C, Li H, Li X, et al. ASK1-dependent endothelial cell activation is critical in ovarian cancer growth and metastasis. JCI Insight. 2017; 2: e91828.
[57]
Aytaç PS, Durmaz I, Houston DR, Çetin-Atalay R, Tozkoparan B. Novel triazolothiadiazines act as potent anticancer agents in liver cancer cells through Akt and ASK-1 proteins. Bioorganic and Medicinal Chemistry. 2016; 24: 858–872.
[58]
Guo X, Namekata K, Kimura A, Harada C, Harada T. ASK1 in neurodegeneration. Advances in Biological Regulation. 2017; 66: 63–71.
[59]
Ji N, Yang Y, Cai CY, Lei ZN, Wang JQ, Gupta P, et al. Selonsertib (GS-4997), an ASK1 inhibitor, antagonizes multidrug resistance in ABCB1-and ABCG2-overexpressing cancer cells. Cancer Letters. 2019; 440-441: 82–93.
[60]
Liu X, Zhang P, Ji K, Zhang J, Yang S, Du B, et al. Cyclin-dependent kinase 5 regulates MAPK/ERK signaling in the skin of mice. Acta Histochemica. 2018; 120: 15–21.
[61]
Ilinca A, Englund E, Samuelsson S, Truvé K, Kafantari E, Martinez-Majander N, et al. MAP3K6 Mutations in a Neurovascular Disease Causing Stroke, Cognitive Impairment, and Tremor. Neurology Genetics. 2021; 7: e548.
[62]
Gaston D, Hansford S, Oliveira C, Nightingale M, Pinheiro H, Macgillivray C, et al. Germline mutations in MAP3K6 are associated with familial gastric cancer. PLoS Genetics. 2014; 10: e1004669.
[63]
Eto N, Miyagishi M, Inagi R, Fujita T, Nangaku M. Mitogen-activated protein 3 kinase 6 mediates angiogenic and tumorigenic effects via vascular endothelial growth factor expression. American Journal of Pathology. 2009; 174: 1553–1563.
[64]
Roh YS, Song J, Seki E. TAK1 regulates hepatic cell survival and carcinogenesis. Journal of Gastroenterology. 2014; 49: 185–194.
[65]
Inokuchi S, Aoyama T, Miura K, Osterreicher CH, Kodama Y, Miyai K, et al. Disruption of TAK1 in hepatocytes causes hepatic injury, inflammation, fibrosis, and carcinogenesis. Proceedings of the National Academy of Sciences of the United States of America. 2010; 107: 844–849.
[66]
Cheng JS, Tsai WL, Liu PF, Goan YG, Lin CW, Tseng HH, et al. The MAP3K7-mTOR Axis Promotes the Proliferation and Malignancy of Hepatocellular Carcinoma Cells. Frontiers in Oncology. 2019; 9: 474.
[67]
Mukhopadhyay H, Lee NY. Multifaceted roles of TAK1 signaling in cancer. Oncogene. 2020; 39: 1402–1413.
[68]
Wen J, Hu Y, Luo K, Yang H, Zhang S, Fu J. Positive transforming growth factor-β activated kinase-1 expression has an unfavorable impact on survival in T3N1-3M0 esophageal squamous cell carcinomas. Annals of Thoracic Surgery. 2013; 95: 285–290.
[69]
Lin P, Niu W, Peng C, Zhang Z, Niu J. The role of TAK1 expression in thyroid cancer. International Journal of Clinical and Experimental Pathology. 2015; 8: 14449–14456.
[70]
Yang Y, Qiu Y, Tang M, Wu Z, Hu W, Chen C. Expression and function of transforming growth factor-β-activated protein kinase 1 in gastric cancer. Molecular Medicine Reports. 2017; 16: 3103–3110.
[71]
Cai PC, Shi L, Liu VW, Tang HW, Liu IJ, Leung TH, et al. Elevated TAK1 augments tumor growth and metastatic capacities of ovarian cancer cells through activation of NF-κB signaling. Oncotarget. 2014; 5: 7549–7562.
[72]
Ninomiya-Tsuji J, Kajino T, Ono K, Ohtomo T, Matsumoto M, Shiina M, et al. A resorcylic acid lactone, 5Z-7-oxozeaenol, prevents inflammation by inhibiting the catalytic activity of TAK1 MAPK kinase kinase. Journal of Biological Chemistry. 2003; 278: 18485–18490.
[73]
Melisi D, Xia Q, Paradiso G, Ling J, Moccia T, Carbone C, et al. Modulation of Pancreatic Cancer Chemoresistance by Inhibition of TAK1. JNCI: Journal of the National Cancer Institute. 2011; 103: 1190–1204.
[74]
Buglio D, Palakurthi S, Byth K, Vega F, Toader D, Saeh J, et al. Essential role of TAK1 in regulating mantle cell lymphoma survival. Blood. 2012; 120: 347–355.
[75]
Totzke J, Gurbani D, Raphemot R, Hughes PF, Bodoor K, Carlson DA, et al. Takinib, a Selective TAK1 Inhibitor, Broadens the Therapeutic Efficacy of TNF-α Inhibition for Cancer and Autoimmune Disease. Cell Chemical Biology. 2017; 24: 1029–1039.e7.
[76]
Tan L, Nomanbhoy T, Gurbani D, Patricelli M, Hunter J, Geng J, et al. Discovery of type II inhibitors of TGFβ-activated kinase 1 (TAK1) and mitogen-activated protein kinase kinase kinase kinase 2 (MAP4K2). Journal of Medicinal Chemistry. 2015; 58: 183–196.
[77]
Tsatsanis C, Patriotis C, Tsichlis PN. Tpl-2 induces IL-2 expression in T-cell lines by triggering multiple signaling pathways that activate NFAT and NF-κB. Oncogene. 1998; 17: 2609–2618.
[78]
Salmeron A, Ahmad TB, Carlile GW, Pappin D, Narsimhan RP, Ley SC. Activation of MEK-1 and SEK-1 by Tpl-2 proto-oncoprotein, a novel MAP kinase kinase kinase. EMBO Journal. 1996; 15: 817–826.
[79]
Patriotis C, Makris A, Chernoff J, Tsichlis PN. Tpl-2 acts in concert with Ras and Raf-1 to activate mitogen-activated protein kinase. Proceedings of the National Academy of Sciences of the United States of America. 1994; 91: 9755–9759.
[80]
Lin X, Cunningham ET Jr, Mu Y, Geleziunas R, Greene WC. The proto-oncogene Cot kinase participates in CD3/CD28 induction of NF-kappaB acting through the NF-kappaB-inducing kinase and IkappaB kinases. Immunity. 1999; 10: 271–280.
[81]
Gkirtzimanaki K, Gkouskou KK, Oleksiewicz U, Nikolaidis G, Vyrla D, Liontos M, et al. TPL2 kinase is a suppressor of lung carcinogenesis. Proceedings of the National Academy of Sciences of the United States of America. 2013; 110: E1470–E1479.
[82]
Decicco-Skinner KL, Jung SA, Tabib T, Gwilliam JC, Alexander H, Goodheart SE, et al. Tpl2 knockout keratinocytes have increased biomarkers for invasion and metastasis. Carcinogenesis. 2013; 34: 2789–2798.
[83]
Tunca B, Tezcan G, Cecener G, Egeli U, Zorluoglu A, Yilmazlar T, et al. Overexpression of CK20, MAP3K8 and EIF5A correlates with poor prognosis in early-onset colorectal cancer patients. Journal of Cancer Research and Clinical Oncology. 2013; 139: 691–702.
[84]
Sourvinos G, Tsatsanis C, Spandidos DA. Overexpression of the Tpl-2/Cot oncogene in human breast cancer. Oncogene. 1999; 18: 4968–4973.
[85]
Lee HW, Cho HJ, Lee SJ, Song HJ, Cho HJ, Park MC, et al. Tpl2 induces castration resistant prostate cancer progression and metastasis. International Journal of Cancer. 2015; 136: 2065–2077.
[86]
Lee JH, Jeong JH, Kim TH, Kim SY, Kim KE, Seong JK, et al. Induction of squamous cell carcinoma after MAP3K8 overexpression in murine salivry gland epithelial cells. Head and Neck. 2019; 41: 924–929.
[87]
Gruosso T, Garnier C, Abelanet S, Kieffer Y, Lemesre V, Bellanger D, et al. MAP3K8/TPL-2/COT is a potential predictive marker for MEK inhibitor treatment in high-grade serous ovarian carcinomas. Nature Communications. 2015; 6: 8583.
[88]
Gallo KA, Johnson GL. Mixed-lineage kinase control of JNK and p38 MAPK pathways. Nature Reviews Molecular Cell Biology. 2002; 3: 663–672.
[89]
Xia J, Cao T, Ma C, Shi Y, Sun Y, Wang ZP, et al. miR-7 Suppresses Tumor Progression by Directly Targeting MAP3K9 in Pancreatic Cancer. Molecular Therapy-Nucleic Acids. 2018; 13: 121–132.
[90]
Ren Q, Xiao X, Leng X, Zhang Q, Zhou X, Ren Z, et al. MicroRNA-361-5p induces hepatocellular carcinoma cell apoptosis and enhances drug sensitivity by targeting MAP3K9. Experimental and Therapeutic Medicine. 2021; 21: 574.
[91]
Abdeyrim A, Cheng X, Lian M, Tan Y. miR‑490‑5p regulates the proliferation, migration, invasion and epithelial‑mesenchymal transition of pharyngolaryngeal cancer cells by targeting mitogen‑activated protein kinase kinasekinase 9. International Journal of Molecular Medicine. 2019; 44: 240–252.
[92]
Fawdar S, Trotter EW, Li Y, Stephenson NL, Hanke F, Marusiak AA, et al. Targeted genetic dependency screen facilitates identification of actionable mutations in FGFR4, MAP3K9, and PAK5 in lung cancer. Proceedings of the National Academy of Sciences of the United States of America. 2013; 110: 12426–12431.
[93]
Marusiak AA, Edwards ZC, Hugo W, Trotter EW, Girotti MR, Stephenson NL, et al. Mixed lineage kinases activate MEK independently of RAF to mediate resistance to RAF inhibitors. Nature Communications. 2014; 5: 3901.
[94]
Stark MS, Woods SL, Gartside MG, Bonazzi VF, Dutton-Regester K, Aoude LG, et al. Frequent somatic mutations in MAP3K5 and MAP3K9 in metastatic melanoma identified by exome sequencing. Nature Genetics. 2012; 44: 165–169.
[95]
Sapkota GP. The TGFβ-induced phosphorylation and activation of p38 mitogen-activated protein kinase is mediated by MAP3K4 and MAP3K10 but not TAK1. Open Biology. 2013; 3: 130067.
[96]
Zhu J, Chen T, Yang L, Li Z, Wong MM, Zheng X, et al. Regulation of microRNA-155 in atherosclerotic inflammatory responses by targeting MAP3K10. PloS ONE. 2012; 7: e46551.
[97]
Wang C, Zhang X, Zhang C, Zhai F, Li Y, Huang Z. MicroRNA-155 targets MAP3K10 and regulates osteosarcoma cell growth. Pathology, Research and Practice. 2017; 213: 389–393.
[98]
Luo W, Zhang H, Liang X, Xia R, Deng H, Yi Q, et al. DNA methylation‑regulated miR‑155‑5p depresses sensitivity of esophageal carcinoma cells to radiation and multiple chemotherapeutic drugs via suppression of MAP3K10. Oncology Reports. 2020; 43: 1692–1704.
[99]
Zhou M, Gao Y, Wang M, Guo X, Li X, Zhu F, et al. MiR-146b-3p regulates proliferation of pancreatic cancer cells with stem cell-like properties by targeting MAP3K10. Journal of Cancer. 2021; 12: 3726–3740.
[100]
Swenson-Fields KI, Sandquist JC, Rossol-Allison J, Blat IC, Wennerberg K, Burridge K, et al. MLK3 limits activated Galphaq signaling to Rho by binding to p63RhoGEF. Molecular Cell. 2008; 32: 43–56.
[101]
Chen J, Gallo KA. MLK3 regulates paxillin phosphorylation in chemokine-mediated breast cancer cell migration and invasion to drive metastasis. Cancer Research. 2012; 72: 4130–4140.
[102]
Chen J, Miller EM, Gallo KA. MLK3 is critical for breast cancer cell migration and promotes a malignant phenotype in mammary epithelial cells. Oncogene. 2010; 29: 4399–4411.
[103]
Rattanasinchai C, Gallo KA. MLK3 Signaling in Cancer Invasion. Cancers. 2016; 8: 51.
[104]
Hazan RB, Qiao R, Keren R, Badano I, Suyama K. Cadherin switch in tumor progression. Annals of the New York Academy of Sciences. 2004; 1014: 155–163.
[105]
Ma L, Cheng Y, Zeng J. MLK3 silence induces cervical cancer cell apoptosis via the Notch-1/autophagy network. Clinical and Experimental Pharmacology and Physiology. 2019; 46: 854–860.
[106]
Li Y, Wang D, Li X, Shao Y, He Y, Yu H, et al. MiR-199a-5p suppresses non-small cell lung cancer via targeting MAP3K11. Journal of Cancer. 2029; 10: 2472–2479.
[107]
Byrnes KA, Phatak P, Mansour D, Xiao L, Zou T, Rao JN, et al. Overexpression of miR-199a-5p decreases esophageal cancer cell proliferation through repression of mitogen-activated protein kinase kinase kinase-11 (MAP3K11). Oncotarget. 2016; 7: 8756–8770.
[108]
Wang J, Deng B, Liu Q, Huang Y, Chen W, Li J, et al. Pyroptosis and ferroptosis induced by mixed lineage kinase 3 (MLK3) signaling in cardiomyocytes are essential for myocardial fibrosis in response to pressure overload. Cell Death and Disease. 2020; 11: 574.
[109]
Yu J, Feng Y, Wang Y, An R. Aryl hydrocarbon receptor enhances the expression of miR-150-5p to suppress in prostate cancer progression by regulating MAP3K12. Archives of Biochemistry and Biophysics. 2018; 654: 47–54.
[110]
Silveira VS, Scrideli CA, Moreno DA, Yunes JA, Queiroz RGP, Toledo SC, et al. Gene expression pattern contributing to prognostic factors in childhood acute lymphoblastic leukemia. Leukemia and Lymphoma. 2013; 54: 310–314.
[111]
Watkins TA, Wang B, Huntwork-Rodriguez S, Yang J, Jiang Z, Eastham-Anderson J, et al. DLK initiates a transcriptional program that couples apoptotic and regenerative responses to axonal injury. Proceedings of the National Academy of Sciences of the United States of America. 2013; 110: 4039–4044.
[112]
Patel S, Cohen F, Dean BJ, De La Torre K, Deshmukh G, Estrada AA, et al. Discovery of dual leucine zipper kinase (DLK, MAP3K12) inhibitors with activity in neurodegeneration models. Journal of Medicinal Chemistry. 2015; 58: 401–418.
[113]
Huntwork-Rodriguez S, Wang B, Watkins T, Ghosh AS, Pozniak CD, Bustos D, et al. JNK-mediated phosphorylation of DLK suppresses its ubiquitination to promote neuronal apoptosis. Journal of Cell Biology. 2013; 202: 747–763.
[114]
Hirai S, Kawaguchi A, Suenaga J, Ono M, Cui DF, Ohno S. Expression of MUK/DLK/ZPK, an activator of the JNK pathway, in the nervous systems of the developing mouse embryo. Gene Expression Patterns. 2005; 5: 517–523.
[115]
Ferraris D, Yang Z, Welsbie D. Dual leucine zipper kinase as a therapeutic target for neurodegenerative conditions. Future Medicinal Chemistry. 2013; 5: 1923–1934.
[116]
Sengupta Ghosh A, Wang B, Pozniak CD, Chen M, Watts RJ, Lewcock JW. DLK induces developmental neuronal degeneration via selective regulation of proapoptotic JNK activity. Journal of Cell Biology. 2011; 194: 751–764.
[117]
Ye M, Li D, Yang J, Xie J, Yu F, Ma Y, et al. MicroRNA-130a Targets MAP3K12 to Modulate Diabetic Endothelial Progenitor Cell Function. Cellular Physiology and Biochemistry. 2015; 36: 712–726.
[118]
Masaki M, Ikeda A, Shiraki E, Oka S, Kawasaki T. Mixed lineage kinase LZK and antioxidant protein-1 activate NF-kappaB synergistically. European Journal of Biochemistry. 2003; 270: 76–83.
[119]
Ikeda A, Hasegawa K, Masaki M, Moriguchi T, Nishida E, Kozutsumi Y, et al. Mixed lineage kinase LZK forms a functional signaling complex with JIP-1, a scaffold protein of the c-Jun NH(2)-terminal kinase pathway. Journal of Biochemistry. 2001; 130: 773–781.
[120]
Han H, Chen Y, Cheng L, Prochownik EV, Li Y. microRNA-206 impairs c-Myc-driven cancer in a synthetic lethal manner by directly inhibiting MAP3K13. Oncotarget. 2016; 7: 16409–16419.
[121]
Edwards ZC, Trotter EW, Torres-Ayuso P, Chapman P, Wood HM, Nyswaner K, et al. Survival of Head and Neck Cancer Cells Relies upon LZK Kinase-Mediated Stabilization of Mutant p53. Cancer Research. 2017; 77: 4961–4972.
[122]
Fu D, Ren Y, Wang C, Yu L, Yu R. LINC01287 facilitates proliferation, migration, invasion and EMT of colon cancer cells via miR-4500/MAP3K13 pathway. BMC Cancer. 2021; 21: 782.
[123]
Chen M, Geoffroy CG, Wong HN, Tress O, Nguyen MT, Holzman LB, et al. Leucine Zipper-bearing Kinase promotes axon growth in mammalian central nervous system neurons. Scientific Reports. 2016; 6: 31482.
[124]
Rahal R, Frick M, Romero R, Korn JM, Kridel R, Chan FC, et al. Pharmacological and genomic profiling identifies NF-κB-targeted treatment strategies for mantle cell lymphoma. Nature Medicine. 2014; 20: 87–92.
[125]
Hamdan TA, Bhat H, Cham LB, Adomati T, Lang J, Li F, et al. Map3k14 as a Regulator of Innate and Adaptive Immune Response during Acute Viral Infection. Pathogens. 2020; 9: 96.
[126]
Schlechter N, Glanzmann B, Hoal EG, Schoeman M, Petersen B, Franke A, et al. Exome Sequencing Identifies a Novel MAP3K14 Mutation in Recessive Atypical Combined Immunodeficiency. Frontiers in Immunology. 2019; 8: 1624.
[127]
Chen Z, Kong H, Cai Z, Chen K, Wu B, Li H, et al. Identification of MAP3K15 as a potential prognostic biomarker and correlation with immune infiltrates in osteosarcoma. Annals of Translational Medicine. 2021; 9: 1179.
[128]
Wu Y, Huang J, Ivan C, Sun Y, Ma S, Mangala LS, et al. MEK inhibition overcomes resistance to EphA2-targeted therapy in uterine cancer. Gynecologic Oncology. 2021; 163: 181–190.
[129]
Naguro I, Umeda T, Kobayashi Y, Maruyama J, Hattori K, Shimizu Y, et al. ASK3 responds to osmotic stress and regulates blood pressure by suppressing WNK1-SPAK/OSR1 signaling in the kidney. Nature Communications. 2012; 3: 1285.
[130]
Dulovic-Mahlow M, Trinh J, Kandaswamy KK, Braathen GJ, Di Donato N, Rahikkala E, et al. De Novo Variants in TAOK1 Cause Neurodevelopmental Disorders. American Journal of Human Genetics. 2019; 1005: 213–220.
[131]
van Woerden GM, Bos M, de Konink C, Distel B, Avagliano Trezza R, Shur NE, et al. TAOK1 is associated with neurodevelopmental disorder and essential for neuronal maturation and cortical development. Human Mutation. 2021; 42: 445–459.
[132]
Zhu L, Yu Q, Gao P, Liu Q, Luo X, Jiang G, et al. TAOK1 positively regulates TLR4-induced inflammatory responses by promoting ERK1/2 activation in macrophages. Molecular Immunology. 2020; 122: 124–131.
[133]
Nourbakhsh K, Ferreccio AA, Bernard MJ, Yadav S. TAOK2 is an ER-localized kinase that catalyzes the dynamic tethering of ER to microtubules. Developmental Cell. 2021; 56: 3321–3333.e5.
[134]
Richter M, Murtaza N, Scharrenberg R, White SH, Johanns O, Walker S, et al. Altered TAOK2 activity causes autism-related neurodevelopmental and cognitive abnormalities through RhoA signaling. Molecular Psychiatry. 2019; 24: 1329–1350.
[135]
Kapfhamer D, Taylor S, Zou ME, Lim JP, Kharazia V, Heberlein U. Taok2 controls behavioral response to ethanol in mice. Genes, Brain, and Behavior. 2013; 12: 87–97.
[136]
Li Z, Oh H, Cung M, Marquez SJ, Sun J, Hammad H, et al. TAOK3 is a MAP3K contributing to osteoblast differentiation and skeletal mineralization. Biochemical and Biophysical Research Communications. 2020; 531: 497–502.
[137]
Xia Y, Caputo M, Cansby E, Anand SK, Sütt S, Henricsson M, et al. STE20-type kinase TAOK3 regulates hepatic lipid partitioning. Molecular Metabolism. 2021; 54: 101353.
[138]
Ormonde JVS, Li Z, Stegen C, Madrenas J. TAOK3 Regulates Canonical TCR Signaling by Preventing Early SHP-1-Mediated Inactivation of LCK. Journal of Immunology. 2018; 201: 3431–3442.
[139]
Ormonde JVS, Nie Y, Madrenas J. TAOK3, a Regulator of LCK-SHP-1 Crosstalk during TCR Signaling. Critical Reviews in Immunology. 2019; 39: 59–81.
[140]
Bian Y, Teper Y, Mathews Griner LA, et al. Target Deconvolution of a Multikinase Inhibitor with Antimetastatic Properties Identifies TAOK3 as a Key Contributor to a Cancer Stem Cell–Like Phenotype. Molecular Cancer Therapeutics. 2019; 18: 2097–2110.
[141]
Lai TC, Fang CY, Jan YH, Hsieh HL, Yang YF, Liu CY, et al. Kinase shRNA screening reveals that TAOK3 enhances microtubule-targeted drug resistance of breast cancer cells via the NF-κB signaling pathway. Cell Communication and Signaling. 2020; 18: 164.
[142]
Boehme SA, Franz-Bacon K, Ludka J, DiTirro DN, Ly TW, Bacon KB. MAP3K19 Is Overexpressed in COPD and Is a Central Mediator of Cigarette Smoke-Induced Pulmonary Inflammation and Lower Airway Destruction. PLoS ONE. 2016; 11: e0167169.
[143]
Boehme SA, Franz-Bacon K, DiTirro DN, Ly TW, Bacon KB. MAP3K19 is a Novel Regulator of TGF-β Signaling That Impacts Bleomycin-Induced Lung Injury and Pulmonary Fibrosis. PLoS ONE. 2016; 11: e0154874.
[144]
Hoang VT, Nyswaner K, Torres-Ayuso P, Brognard J. The protein kinase MAP3K19 phosphorylates MAP2Ks and thereby activates ERK and JNK kinases and increases viability of KRAS-mutant lung cancer cells. Journal of Biological Chemistry. 2020; 295: 8470–8479.
[145]
Jones IC, Espindola MS, Narayanan R, Coelho AL, Habiel DM, Boehme SA, et al. Targeting MAP3K19 prevents human lung myofibroblast activation both in vitro and in a humanized SCID model of idiopathic pulmonary fibrosis. Scientific Reports. 2019; 9: 19796.
[146]
Fu CY, Chen MC, Tseng YS, Chen MC, Zhou Z, Yang JJ, et al. Fisetin activates Hippo pathway and JNK/ERK/AP-1 signaling to inhibit proliferation and induce apoptosis of human osteosarcoma cells via ZAK overexpression. Environmental Toxicology. 2019; 34: 902–911.
[147]
Quan Y, Zhang Y, Lin W, Shen Z, Wu S, Zhu C, et al. Knockdown of long non-coding RNA MAP3K20 antisense RNA 1 inhibits gastric cancer growth through epigenetically regulating miR-375. Biochemical and Biophysical Research Communications. 2018; 497: 527–534.
[148]
Fu CY, Lay IS, Shibu MA, Tseng YS, Kuo WW, Yang JJ, et al. Selective Activation of ZAK β Expression by 3-Hydroxy-2-Phenylchromone Inhibits Human Osteosarcoma Cells and Triggers Apoptosis via JNK Activation. International Journal of Molecular Sciences. 2020; 21: 3366.
[149]
Rey C, Faustin B, Mahouche I, Ruggieri R, Brulard C, Ichas F, et al. The MAP3K ZAK, a novel modulator of ERK-dependent migration, is upregulated in colorectal cancer. Oncogene. 2017; 35: 3190–3200.
[150]
Wang W, Zhou C, Tang H, Yu Y, Zhang Q. Combined Analysis of DNA Methylome and Transcriptome Reveal Novel Candidate Genes Related to Porcine Escherichia coli F4ab/ac-Induced Diarrhea. Frontiers in Cellular and Infection Microbiology. 2020; 10: 250.
[151]
Mao K, Zhang M, Cao J, Zhao X, Gao L, Fu L, et al. Coding Variants are Relevant to the Expression of Obesity-Related Genes for Pediatric Adiposity. Obesity. 2021; 29: 194–203.
[152]
Banos G, Lindsay V, Desta TT, Bettridge J, Sanchez-Molano E, Vallejo-Trujillo A, et al. Integrating Genetic and Genomic Analyses of Combined Health Data Across Ecotypes to Improve Disease Resistance in Indigenous African Chickens. Frontiers in Genetics. 2020; 11: 543890.
[153]
Martini M, Russo M, Lamba S, Vitiello E, Crowley EH, Sassi F, et al. Mixed lineage kinase MLK4 is activated in colorectal cancers where it synergistically cooperates with activated RAS signaling in driving tumorigenesis. Cancer Research. 2013; 73: 1912–1921.
[154]
Marusiak AA, Stephenson NL, Baik H, Trotter EW, Li Y, Blyth K, et al. Recurrent MLK4 Loss-of-Function Mutations Suppress JNK Signaling to Promote Colon Tumorigenesis. Cancer Research. 2016; 76: 724–735.
[155]
Li Y, Zuo H, Wang H, Hu A. Decrease of MLK4 prevents hepatocellular carcinoma (HCC) through reducing metastasis and inducing apoptosis regulated by ROS/MAPKs signaling. Biomedicine and Pharmacotherapy. 2019; 116: 108749.
[156]
Kim SH, Ezhilarasan R, Phillips E, Gallego-Perez D, Sparks A, Taylor D, et al. Serine/Threonine Kinase MLK4 Determines Mesenchymal Identity in Glioma Stem Cells in an NF-κB-dependent Manner. Cancer Cell. 2016; 29: 201–213.
[157]
Blessing NA, Kasturirangan S, Zink EM, Schroyer AL, Chadee DN. Osmotic and heat stress-dependent regulation of MLK4β and MLK3 by the CHIP E3 ligase in ovarian cancer cells. Cellular Signalling. 2017; 39: 66–73.
[158]
Brandl L, Horst D, Grünewald TGP, Mayerle J, Sendelhofert A, Neumann J, et al. Clinical Evidence on the Interaction between MLK4, KRAS and Microsatellite Instability to Determine the Prognosis of Early-Stage Colorectal Carcinoma. Cellular Physiology and Biochemistry. 2019; 53: 820–831.
[159]
Shan J, Chouchane A, Mokrab Y, Saad M, Boujassoum S, Sayaman RW, et al. Genetic Variation in CCL5 Signaling Genes and Triple Negative Breast Cancer: Susceptibility and Prognosis Implications. Frontiers in Oncology. 2019; 9: 1328.
[160]
Marusiak AA, Prelowska MK, Mehlich D, Lazniewski M, Kaminska K, Gorczynski A, et al. Upregulation of MLK4 promotes migratory and invasive potential of breast cancer cells. Oncogene. 2019; 38: 2860–2875.
[161]
Nguyen K, Yousefi H, Cheng T, Magrath J, Hartono AB, Alzoubi M, et al. Expression of Novel Kinase MAP3K19 in Various Cancers and Survival Correlations. Frontiers in Bioscience (Landmark Edition). 2022;
[162]
Fedorov O, Müller S, Knapp S. The (un)targeted cancer kinome. Nature Chemical Biology. 2010; 6: 166–169.
[163]
Knapp S, Arruda P, Blagg J, Burley S, Drewry DH, Edwards A, et al. A public-private partnership to unlock the untargeted kinome. Nature Chemical Biology. 2013; 9: 3–6.
Share
Back to top