Next Article in Journal
Assessment of Automated Flow Cytometry Data Analysis Tools within Cell and Gene Therapy Manufacturing
Next Article in Special Issue
A Bioengineered In Vitro Model to Assess AAV-Based Gene Therapies for Cyclic GMP-Related Disorders
Previous Article in Journal
HNF1A Mutations and Beta Cell Dysfunction in Diabetes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Oncogenic Pathways in Neurodegenerative Diseases

by
Luis Varela
1,* and
Maria E. R. Garcia-Rendueles
2,*
1
Yale Center for Molecular and Systems Metabolism, Department of Comparative Medicine, School of Medicine, Yale University, 310 Cedar St. BML 330, New Haven, CT 06520, USA
2
Precision Nutrition and Cancer Program, IMDEA Food Institute, Campus Excelencia Internacional UAM+CSIC, 28049 Madrid, Spain
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(6), 3223; https://doi.org/10.3390/ijms23063223
Submission received: 28 February 2022 / Revised: 14 March 2022 / Accepted: 15 March 2022 / Published: 17 March 2022
(This article belongs to the Special Issue Genetics of Health and Disease)

Abstract

:
Cancer and neurodegenerative diseases are two of the leading causes of premature death in modern societies. Their incidence continues to increase, and in the near future, it is believed that cancer will kill more than 20 million people per year, and neurodegenerative diseases, due to the aging of the world population, will double their prevalence. The onset and the progression of both diseases are defined by dysregulation of the same molecular signaling pathways. However, whereas in cancer, these alterations lead to cell survival and proliferation, neurodegenerative diseases trigger cell death and apoptosis. The study of the mechanisms underlying these opposite final responses to the same molecular trigger is key to providing a better understanding of the diseases and finding more accurate treatments. Here, we review the ten most common signaling pathways altered in cancer and analyze them in the context of different neurodegenerative diseases such as Alzheimer’s (AD), Parkinson’s (PD), and Huntington’s (HD) diseases.

1. Introduction

Cancer and neurogenerative diseases are two of the leading causes of death in modern societies, and despite all the efforts made in understanding their onset and development, their prevalence continues to increase dramatically. Cancer, together with cardiovascular disease, is known to be the main cause of premature death [1]. In the last years, its incidence has risen rapidly. It reached an estimated 20 million new cases worldwide within 2020, and it is believed that in the next two decades, the incidence of cancer will increase by 50%. Cancer was also the cause of 10 million deaths last year [2]. In line with this, neurodegenerative disorders are a group of age-related diseases [3] that affect millions of people worldwide. They have become an important public health burden, with increasing incidence and mortality and an associated rise in healthcare costs [4,5]. The fact that aging is the major risk for neurodegeneration, and together with the expectations that the aged population will exceed the number of young individuals in the next decades, makes neurodegenerative diseases one of the most important threats to the well-being of individuals and society.
Although the causes and consequences of the different neurodegenerative diseases are various, their common clinical features are marked by a progressive loss of cognitive function, defective motor coordination, and increased pain triggered by, in all cases, a loss of specific neuronal populations (Table 1) [6]. Alzheimer’s (AD) and Parkinson’s (PD) diseases are the two most prevalent neurodegenerative disorders, and they are characterized by the aberrant accumulation of aggregates, Amyloid β (Aβ) in senile plaques and Tau in neurofibrillary tangles in AD, and α-synuclein in Lewis bodies in PD [7,8]. Despite the hallmarks of AD and PD being identified, the mechanisms underlying their development remain far from completely understood. Altered oxidative stress, cell cycle activation, and inflammation, among others, are the stimuli that trigger neurodegeneration [9,10]. Although rare, accounting for 5% of all the cases, mutations in specific genes are the cause of familial AD and PD. APP, APOE, PARKIN, and PINK1 have been identified as causal genes of AD and PD [11,12]. On the contrary, Huntington’s disease (HD) is a hereditary disease caused by the mutation of HTT [13].
In cancer, cells acquired the ability to divide and growth uncontrollably [33,34,35]. Unlike normal cells, cancer cells do not respond to the controlling signals mainly due to molecular alterations in specific genes associated with signalling pathways. The complexity is since, all these signalling routes are connected forming an intricated signalling network, thus oncogenic mutations can affect proteins implicated in several signalling pathways, such as Notch-Wnt-TGFb-Hippo pathways. Moreover, there is considerable variation in the genes and pathways altered across different tumor types and individual tumor samples.
Although the mechanisms underlying cancer and neurodegenerative disorders are different, the onset and progression of both diseases share the same molecular signaling pathways. In this review, we provide a summary of the molecular alterations implicated in neurodegenerative diseases, based on the ten canonical signal pathways most altered in cancer [36]. The objective is to understand the role of critical cancer pathways in neurodegenerative diseases.

2. Oncogenic Signaling Pathways

2.1. Hippo Pathway

First discovered as a regulator of organ size, Hippo signalling is involved in many different processes, such as mechanotransduction, homeostasis, cellular differentiation, and tissue regeneration, among others [37,38,39]. Briefly, the canonical Hippo pathway induces the activation of MST1/2 (mammalian sterile 20-like kinases 1 and 2) which, through the phosphorylation of LATS1/2 (large tumor suppressors 1 and 2), phosphorylates YAP/TAZ. Phosphorylated YAP (Yes-associated protein) is retained in the cytoplasm and marked for proteasomal degradation. Non-phosphorylated YAP is translocated to the nucleus, where it complexes with different transcription factors to initiate the transcription of genes involved in cell proliferation and survival [39,40]. YAP overexpression and nuclear localization have been described in different cancers due to the inactivation of the Hippo pathway or the constitutive activation of YAP. This aberrant location of YAP drives the transcription of genes involved in metastasis, pro-tumoral microenvironment, or anti-apoptosis [41,42]. Mutations in Hippo components are rare in cancer [43,44] ], NF2 being the most mutated one. Germline heterozygotic mutations in NF2 cause neurofibromatosis type 2 that predisposes to tumors of the nervous system such as schwannomas, meningiomas, and ependymomas [45,46]. YAP nuclear localization and activation are the principal cause of tumorigenesis and drug resistance [47,48].
The Hippo pathway has been well-studied in the developing brain (reviewed in [49,50]), but its relevance in the adult brain has emerged recently. Hippo pathway components have been suggested as early markers of degenerative diseases within the developing brain [51]. Different integrated analysis studies of postmortem brains revealed the downregulation in Hippo pathway-related genes in various brain areas of AD patients [52,53]. In line with these findings, in AD mouse models, YAP mRNA expression is downregulated in the earlier stages of the disease. The subcellular location of YAP was found altered in postmortem brains of MCI and AD patients [54]. The appearance of Aβ aggregates sequestrates YAP in the cytoplasm of cortical neurons, reducing the accumulation of YAP in the nucleus of these neurons. The mouse models of AD 5xFAD and amyloid precursor protein (APP) knock-in [54] present YAP cytoplasmatic location even before the onset of cognitive impairments. Interestingly, overexpression of YAP, by administration of AAV-YAPdeltaC61 into the cerebrospinal fluid (CSF) space, increased the levels of nuclear YAP, decreased extracellular Aβ plaques, and restored different behavioral parameters of 5xFAD mice to levels similar to control mice [54].
Changes in YAP location are not exclusive of AD; it also has been found in postmortem brains of Huntington’s disease patients as reported [55,56]. In cortical neurons of HD patients, YAP is localized mainly off the nucleus. Mouse models revealed increased levels of total YAP and phosphorylated YAP in the striatum and cortex [55]. Interestingly, cytoplastic YAP localization in the neurons of AD and HD patients has been linked to a new mechanism of necrosis, TRIAD (TEAD-YAP dependent necrosis) [56]. TRIAD, characterized by ER enlargement, has been found in different mouse models of neurodegenerative diseases. Sequestration of YAP in the cytoplasm seems to drive the appearance of ER ballooning. Abnormal morphology of the ER is reversed by specific overexpression of YAP [54].
Although YAP has lately gained relevance as the main effector of the Hippo signaling in the onset of neurodegenerative disorders, other components of the pathway, such as MST1 and LATS1/2, have been also identified and linked to the progression of different CNS diseases [57,58]. For example, higher levels of phospho-MST1 were reported in the motor neurons of the spinal cord in both ALS patients and animal models [58]. In PD, MST1 is involved in dopaminergic neuronal loss. Activated MST1 phosphorylates UNC5B, a pro-apoptotic netrin family receptor, causes motor dysfunctions and reduced dopaminergic cell counts in the substantia nigra (SN) [57]. Similar to YAP, MST1 was found overexpressed in the postmortem brains of HD patients [55].

2.2. Notch Pathway

Notch is a conserved pathway responsible for a wide range of physiological roles, including self-renewal, differentiation, angiogenesis, and proliferation [59,60]. Notch activity is reported to have complex and context-dependent effects [61]. The canonical Notch pathway includes the activation of the Notch family receptors (Notch 1, 2, 3, and 4) by the binding of DSL ligands that induce the cleavage of Notch in NECD (Notch extracellular domain) and NICD (Notch intracellular domain) [62,63]. NICD is translocated into the nucleus where it forms a complex with Mastermind-like (MAML) and other co-activators to stimulate the transcription of Notch target genes. Mutations in Notch receptors have been found in T-cell acute lymphoblastic leukemia, breast cancer, and adenoid cystic carcinoma [64,65,66,67].
The Notch signaling pathway regulates neurogenesis, neural maturation, and synaptic plasticity in the brain [68,69,70]. Moreover, in the adult brain, Notch has been shown to play an important role in the formation of Aβ plaques [70]. Notch colocalizes with presenilins (PSs), the catalytic component of γ-secretase that cleaves APP and induces the aggregation of Aβ [71,72]. Different studies have detected abnormal expression of Notch in the postmortem brains of AD patients [73,74]. Findings from the 1990s pointed at neurons as the key players of this aberrant expression of Notch [73]. The first studies identified aberrant expression of Notch in the hippocampus of AD patients and pointed at neurons from different areas as the main sites of expression of Notch. Recently, a series of works have focused in more detail on the location of Notch in AD patients. In a very elegant study, Brai et al. [75] described that the increased expression of Notch in the postmortem brains of AD patients was due to their aggregation in plaque-like structures. Neurons from different cortical and hippocampal areas presented lesser expression of Notch due to the specific decrease of the extracellular domain of Notch levels. The authors also showed that NECD accumulated in Aβ plaques in brains of AD patients. Interestingly, all the Notch- and Aβ-positive plaques were invaded by microglia and astrocytes, suggesting a potential involvement of these cells in pro-inflammatory response to Notch delocalization [75]. Moreover, AD patients presented less Notch expression in the CSF than healthy patients [70]. Notch accumulation in plaque-like structure in the parenchyma reduced the filtration to the CSF.
Notch ligands were also involved in the onset and development of different neurodegenerative diseases. Among others, alterations in Jagged1 were observed in AD patients [76]. Like the specific reduction of Notch in hippocampal and cortical neurons, AD patients presented fewer Jagged1 expression levels. The generation of a mouse model with a deleted expression of Jagged in neurons showed a potential role of Notch ligand in memory loss. Specifically, this animal model has a reduced expression of Notch in hippocampal neurons and presented an impaired spatial memory similar to the observed in AD patients [29].
Although to a lesser extent, Notch signaling is also related to other neurodegenerative diseases, such as PD and Down syndrome [77,78].

2.3. Nrf2 Pathway

Nrf2 (Nuclear factor-erythroid factor 2-related factor 2) holds key physiological functions in homeostasis maintenance and cell proliferation. It is a master regulator of redox balance and antioxidant-related activity [79]. Recently, its role in metabolic reprogramming was described [80]. Under basal conditions, Nrf2 is sequestered with Keap1 (Kelch-like ECH-associated protein 1) and leads to CUL3-mediated ubiquitination followed by proteasome degradation [81]. Under upstream signals, such as oxidative stress, Nrf2 dissociates from Keap1, translocates to the nucleus, complexes with ARE and other transcription factors, and induces the transcription of detoxification, antioxidant, metabolism, or proliferative genes [82,83]. Constitutive Nrf2 nuclear localization and hyperactivation correlates with cancer progression and chemoresistance, in glioblastoma, lung, hepatocellular carcinoma, cervical, and pancreatic cancer [84]. Nrf2 activation inhibits apoptosis and increases proliferation and invasion [85,86].
Nrf2 has been widely studied within the CNS [87,88,89]. Despite the different neurodegenerative diseases having diverse causes and consequences, oxidative stress is a common pathogenic mechanism in such disorders. In this sense, Nrf2 emerges as a crucial factor in oxidative damage response found in the early stages of AD and PD [90,91]. Nrf2 antioxidant effects have been proposed as a therapeutical target for the treatment of neurological disorders. Nrf2’s role in AD and PD was first reported, in the study of postmortem brains of AD and PD patients [92]. There are differences in the Nrf2 subcellular location between AD and PD brains. Whereas in hippocampal neurons from AD patients the Nrf2 staining was mainly cytoplasmatic, in the SN of PD patients, dopaminergic cells exhibited a strong nuclear location of Nrf2 [92]. Although the reason for these differences in the subcellular location of Nrf2 remains unknown, one possible explanation given by the authors of the study might settle on the dynamics and the timing of the disease. In situations of enhanced oxidative stress, Nrf2 is translocated to the nucleus, where it triggers the transcription of genes involved in the antioxidant response in neurons [92]. The fact that in AD Nrf2 is found mainly in the cytoplasm is a sign of disrupted acclimation of neurons to the oxidative conditions. On the other hand, it is known that PD produces dopaminergic cell loss. The nuclear location of Nrf2 in the SN of PD patients could probably be seen in the neurons that still maintain proper functions, while those dead dopaminergic cells have no Nrf2 staining.
There are numerous efforts to determine the relevance of Nrf2 in the development of neurodegenerative disorders using transgenic mouse models. AD mouse models (APP/PS1 mouse) showed defective expression of Nrf2 and its downstream targets in the hippocampus and cortex, coinciding with the increase of Aβ aggregates [93,94]. Interestingly, lentivirus-meditated overexpression of Nrf2 in the hippocampus of APP/PS1 mouse improves the learning deficits while reducing the levels of soluble Aβ of this mouse strain [95]. Loss of Nrf2 also exacerbates the effects of other AD mouse models in Aβ deposition and spatial learning and memory [93].
Nrf2 involvement in the progression of neurodegenerative disorders has also been linked to neuroinflammation and autophagy. p62, an autophagy marker, is closely related to the Nrf2 signaling pathway [88,96]. Loss of this function impairs the clearance of Aβ aggregates. p62 accumulates in the cytoplasm in absence of autophagy and interacts with the complex t Nrf2-Keap1 [89,97]. p62 releases Nrf2 from Keap1 and is translocated to the nucleus to induce the transcription of antioxidant enzymes and autophagy-related genes. Interaction between p62 and Nrf2 is a positive feedback loop, where defective autophagy [97] activates the oxidative stress response and autophagy itself. The imbalance of this mechanism has been shown to be relevant in the progression of neurological disorders.

2.4. WNT/β-Catenin Pathway

The Wnt/β-catenin pathway is a highly conserved pathway that regulates key cellular functions including gene stability, differentiation, proliferation, apoptosis, stem cell renewal, and migration [98,99,100]. The canonical pathway consists of WNT proteins binding to frizzled receptors and LRP co-receptors, suppressing the activity of the “β-catenin destruction complex” and free β-catenin. This complex is composed of APC (adenomatous polyposis coli), Axin, CK1α (casein kinase 1α), and GSK-3β (glycogen synthase kinase-3β). β-catenin translocates to the nucleus where it associates with Tcf/Lef inducing the transcription of proliferation genes, such as c-Myc, cyclin D1, or c-JUN [101,102,103]. In the absence of WNT ligand, “β-catenin destruction complex” is active, recruit -TrCP E3 linker (β-transducin repeat-containing protein, an E3 ubiquitin ligase), and subsequently degrade β-catenin via the proteasome. In the cytoplasm, β-catenin could form complex with adherent junctions and promotes cell-to-cell adhesion. This pathway has been found altered in cancer and is involved in initiation, progression, and metastasis, involving CSC (cancer stem cell) activation. APC mutations have been found in 90% of colorectal, 70% of gastric cancers, 50% of liver cancers, and 5% of colorectal [100]. Melanoma, prostate, thyroid, and ovary showed mutations in β-catenin [100]. CTNNB1 mutations are present in 90% of WNT-activated medulloblastomas [104]; mutations in β-catenin, APC and AXIN1 have also frequently been identified in medulloblastoma [105,106,107].
Despite the well-known role of WNT signaling in the developing brain [108,109], where it controls synapse formation or neurogenesis, its function in the mature brain is not fully unraveled. Different studies have shown that several components of the WNT signaling are altered in age-related disorders and have been linked to tau and amyloid pathologies, hallmarks of AD disease [110].
Different players of the WNT pathway are altered in the postmortem brains of AD patients [111]. DKK1, an extracellular ligand of WNT receptors and negative regulator of the pathway, is highly expressed in cortical neurons of the diseased brain [112,113]. LPR6, co-receptor of the WNT signaling, is downregulated in the temporal cortex of AD patients. Interestingly, LRP6 downregulation is associated with a lower expression of β-catenin, and hence, to a lower translocation of this protein to the nucleus, where it activates WNT target genes [110].
Different mouse strains of neurogenerative disease also showed altered expression of WNT components [94,110,112]. Mouse models of amyloid deposition and tau pathology showed increased expression of DKK1, accompanied by lower levels of β-catenin. Interestingly, the important role of WNT pathway in the formation of Aβ plaques has been demonstrated in different mouse models. Loss of WNT signaling exacerbates the amyloid deposition in mouse models of amyloid pathology [113].
In postmortem brains derived from patients suffering from other neurodegenerative disorders, such as Parkinson’s and Huntington’s diseases, scientists have found dysregulation of the WNT pathway. For example, genes regulated by WNT were found downrated in the SN of PD brains [111,113,114]. The mechanism by which WNT affects dopaminergic cell loss in SN of PD patients is linked to its role in synapse formation and cell regeneration.

2.5. TGFβ Pathway

The TGFβ (transforming growth factor β) superfamily of growth factors includes TGFβ, activins, and BMPs. The activated TGFβ ligands interact with type II TGFβ receptors (TβRII), which subsequently recruit and phosphorylate type I TGFβ receptors (TβRI), thereby activating downstream signaling through either the SMAD-dependent canonical pathway or the SMAD-independent non-canonical pathway. The canonical pathway involves R-Smad (Smad2/3) phosphorylation complex with co-Smad (Smad4) to translocate to the nucleus and activate the expression of genes [115,116]. The non-canonical pathway activates other upstream components of different pathways such as tumor necrosis factor (TNF) receptor-associated factor (TRAF) 4 or TRAF6, TGFβ-activated kinase 1 (TAK1), Rho GTPases, mitogen-activated protein kinase (ERK or p38), jun N-terminal kinase (JNK), or nuclear factor-κB (NF-κB) [117,118,119].
In normal cells, the TGFβ pathway regulates key physiological functions in homeostasis, development, tissue, and cell growth [120]. In cancer, TGFβ has opposite roles. While in early stages, TGFβ acts as a tumor suppressor by inducing apoptosis and promoting cell-cycle arrest; in advanced-stage cancers, TGFβ acts as a tumor promoter [120,121,122,123]. Cancer cells escape to growth control by mutations or epigenetic modifications in the components of the TGFβ signaling cascade or by becoming resistant to the suppressive effects of TGFβ signaling [120,121,122,123]. TGFβ turns into an oncogenic factor and induces proliferation, epithelial to mesenchymal transition (EMT), evasion of immune surveillance, angiogenesis, drug resistance, and cancer cell stemness. High levels of TGFβ in patients with breast, lung, colorectal, and thyroid cancer have been described, and it is a prediction for poor prognosis [124,125,126,127,128,129].
The TGFβ superfamily is involved in neuroinflammation and repair after brain injury [130]. In the CNS, the TGFβ pathway components have low expression. Astrocytes are the principal source of TGFβ, while TGFβ receptors are in the neurons of different restricted areas [131]. It has been shown that TGFβ plays an important role in age-dependent diseases. Aging increases TGFβ expression in the brain [132]. Although many works have studied this pathway in the pathology of different neurodegenerative disorders, its exact role is not completely understood. Some studies showed TGFβ’s beneficial role in the onset of AD, PD, and other diseases, and others have reported its detrimental effects. Similar to cancer, TGFβ seems to play a dual role in neurogenerative disorders depending on the specific context [130]. TGFβ pathway has been shown altered in brain, CSF, and blood of human AD, PD, and HD patients [132,133,134,135,136]. In AD patients, TGFβ is decreased in plasma but increased in CSF [132], TβRII expression is reduced in the brain, the phosphorylation of SMAD2/3 and its subcellular location are also altered in the diseased brain [137]. Incongruity has also been found in HD, where some reports described increased plasmatic levels of TGFβ [138], and others found lower levels of TGFβ in blood [133].
In the AD brain, the TGFβ pathway promotes amyloid deposition and is co-expressed with Tau in neurons and tangles [139]. TβRII silencing in cortical and hippocampal neurons decreased TGFβ activity and triggered neurodegeneration. TβRII deficient mice showed a smaller number of neurons but a higher number of astrocytes [131]. The TGFβ role in the pathogenesis of the disease was corroborated by breeding the mice losing TGFβ signaling to mouse models of AD. Interestingly, the combination of both models induced an enhanced accumulation of Aβ plaques due to increased levels of APP found in the neurons of this model [131]. A pharmacological study showed that central administration of TGFβ reduced plaque formation and rescued the Aβ induced cognitive impairment [140]. Other works, however, have described that overexpression of TGFβ induces the onset of amyloid pathology [141,142].
TGFβ seems to be involved in the onset and development of Tau pathology. TGFβ levels have been correlated with NFTs in AD brain. Moreover, Smad2/3 co-expressed with Tau in both neurons and NFTs [137,139].
TGFβ has different implications in the development of PD. It is increased in the brains of PD patients [143]. Several transgenic mouse models losing TGFβ signaling presented a reduced number of dopaminergic cells within the SN [144], suggesting that impairment of the TGFβ signaling increases the risk of PD. Although opposite effects of TGFβ have been described, a recent study shows that loss of TGFβ signaling in neurons shows age-related memory and cognitive deficit and presents an important sign of degeneration in the SN. Overexpression of TβRI in the SN of wild type-mice protected against MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) -induced neurodegeneration and cognitive loss [144].
Although to a lesser extent, a potential role of TGFβ in the cortical neurons of HD patients has been described [145]. TGFβ signaling was also found altered in Pick’s disease and ALS, involving the subcellular location of Smad2/3 [137].

2.6. MYC Pathway

C-Myc belongs to the MYC family that also encompasses N-Myc and L-Myc proteins in mammalian cells. Although highly homologous, they display more tissue-restricted expression [146]. c-Myc heterodimerizes with MAX protein, and the complex binds “E-boxes” with a consensus sequence 5′-CACGTG-3′, enriched in the promoters and enhancers to regulate gene expression [147,148]. c-Myc plays a role as a signal node, so under normal circumstances, its expression is tightly regulated by important pathways, such as Wnt/beta-catenin, Ras/Raf/ERK, and the Ras/PI3K/AKT/GSK-3 pathways. As a transcription factor, c-Myc responds to and integrates these signals into broad changes in gene expression, supporting cell growth, proliferation, apoptosis, energy metabolism with biomass accumulation, and diverse biosynthetic pathways [149,150,151,152,153]. In malignant cells where c-Myc is overexpressed, c-Myc can bind DNA sequences beyond E-boxes [154].
Myc is frequently dysregulated during tumorigenesis and is a central driver in multiple cancers, such as breast cancer [155], liver tumor [156], colorectal carcinoma [157], and prostatic neoplasia [153]. Either high, aberrant, or in combination, Myc expression occurs in >70% of human cancers and is related to poor prognosis and aggressive conditions [158,159,160]. MYC alterations have been found in cancer but were mutually exclusive with PIK3CA, PTEN, APC, or BRAF alterations, suggesting that MYC is a distinct oncogenic driver [161].
Although the characterization of the MYC members has been very extensive in the study of different cancer types, its role in neurodegenerative diseases is still far from being understood. Similar to its functions in cancer progression, MYC has also been tightly linked with cell cycle re-entry in the onset and development of AD and other CNS disorders [162].
Alterations of the different Myc members were found in the brains of AD and HD patients [163,164]. Whereas n-Myc expression is decreased, specifically in AD brains, HD seems to affect only the c-Myc expression in PD brains, there is no difference in the expression pattern of Myc members [164]. A subsequent study analyzes the phosphorylation of c-Myc and its subcellular location in AD. Interestingly, despite no differences being reported in total c-Myc expression in the hippocampus of AD brains, phosphorylation status seems to be altered [164]. Phospho-c-Myc was found in NFT positive neurons and in the vicinity of senile plaques in AD, Pick’s disease, and other neurogenerative disorders [164]. Increased mRNA levels of c-Myc were found in the human brain and a mouse model of AD [165].
Insights into the relevance of c-Myc in the onset of neurodegenerative diseases were first reported by a study that generated a mouse strain expressing c-Myc in neurons [166]. Increased expression of c-Myc triggered neuronal loss in the hippocampus and memory deficits. Moreover, another study showed the role of n-Myc in CNS disorders development. Ablation of NDRG2, n-Myc downstream gene 2, exacerbates the AD-like phenotype in pharmacological and genetic models of AD [167]. NRG2 levels were affected by Aβ in a mouse model of AD [167]. Increasing levels of NRG2 were linked to increased expression of APP and the appearance of Aβ plaques [167].

2.7. MAPK Pathway

The mitogen-activated protein kinase (MAPK) is a complex interconnected signaling cascade that converges in the amplification of key molecules that sustain cell proliferation, growth, and survival processes [168]. The MAPK family consists of mainly four signaling families activated by receptor tyrosine kinases (TRKs): MAPK/ERK, the Big MAP kinase-1 (BMK-1), c-Jun N-terminal kinase (JNK), and p38 signaling families. The canonical pathway involves ligand-bound RTKs that activate RAS (GTPases family HRAS, KRAS, and NRAS) with the consequent activation by phosphorylation of RAF family members (ARAF, BRAF, and CRAF), MEK and ERK, the final effector. Nuclear pERK activates the transcription of survival, proliferation, and differentiation genes [169,170].
The MAPK pathway is one of the most altered pathways in cancer. Braf is altered in 60% of thyroid cancer, 54% of melanoma, and 11% colorectal cancers [36,171,172]. Mutations in BRAFV600E are by far the most identified mutation in human tumors. This mutation results in strong BRAF kinase activation (independent of upstream signal) and constitutively hyperactivation of the MAPK pathway. Approximately 19% of patients with cancer harbor RAS mutations, with KRAS responsible for 75% of that number. Furthermore, 65% of pancreatic cancers harbor an RAS mutation, 47% of colorectal, 30% of melanoma and lung cancers, and 12% of thyroid cancer. KRAS is the main isoform mutated in pancreatic and colorectal, NRAS in melanoma and thyroid carcinoma. Moreover, mutations also occur in the genes coding for the tyrosine kinase receptors (EGFR, c-MET, c-KIT) [36,171,172]. EGFR is frequently altered in glioblastoma: 44% presented EGFR amplification, and 23% harbor EGFR mutations [36,171,172].
The canonical ERK cascade, the p38 and the JNK pathways play an important role in different neurological disorders. In the brain, the MAPK pathway is involved in neurogenesis, gliogenesis, and synapse transmission, which affects cognition and memory formation. Pharmacological and genetic approaches showed that alteration of the different MAPK pathways leads to changes in behavior in terms of cognition and learning, incipient signs of neurological disorders [173].
In human patients with AD, PD, HD, or other neurological disorders, increased levels of activated ERK, p38, and JNK have been found. Higher pMEK and pERK levels were found in AD [174,175,176] and PD [177] brains at different stages. The active form of p38 (phosphorylated) is upregulated in the early stages of AD [178,179]. JNKs were reported to increase not only in different areas of the brains of AD patients but also in their CSF. JNKs play a relevant role in the dopaminergic cell loss characteristic of PD. Furthermore, in several models of AD and PD, the different MAPK signaling pathways were demonstrated to be upregulated [180].
In AD, MAPK pathways are linked to both amyloid and tau pathologies. In fact, in the brains of AD patients, different members of the cascade members were found to co-express in NFTs and senile plaques. Moreover, pharmacological blockade and genetic deletion of pERK, p38 and JNK ameliorate the cognitive impairments in different mouse models of AD [180,181,182,183,184,185]. These restored memory and learning functions are associated with diminished levels of Aβ accumulation. Several reports have linked the expression of APP with the activity of the MAPK pathway. For example, lower amounts of Aβ deposition driven by the loss of p38 in an AD model are associated with a decrease in the β-secretase activity. Another study [185] showed that JNK inhibition also ameliorates working memory, and this is associated with a reduction of plaques in cortical, and hippocampal areas, lower levels of secretase activity, and diminished expression of phosphorylated APP.
ERK, p38, and JNK pathways are involved in the survival of dopaminergic cells in the striatum and the dopaminergic signaling of the brain [186]. It has been shown that pharmacological models of parkinsonism have altered the expression of all the most important MAPK pathways [185,186]. Regarding the genetic ablation of JNK2 protected against the MPTP-induced Parkinson model, this study is the first to show the possible relevance of MAPKs in the development of PD [186]. Moreover, pharmacological blockade of another isoform of JNK (JNK3) mitigates the dopaminergic cell loss induced by MPTP [181]. p38 is involved in the dopaminergic cell loss by responding to dysregulated oxidative stress in these neurons. It was also described that α-synuclein induces the expression of p38, ERK, and JNK in glial cells [187,188]. α-synuclein released by damaged neurons signals to microglia and triggers and pro-inflammatory responses of these glial cells [188].
At the onset and development of HD, MAPK may also play an important role. Like humans, mouse models mimicking the effect of HD [189] presented increased levels of phosphorylated p38 and JNK in the striatum. Different studies in cell cultures suggest that mutant HTT affects MAPK pathways and activates them [190].

2.8. p53 Pathway

p53 is a potent tumor suppressor, and it is considered the “guardian of the genome” to prevent the accumulation of oncogenic mutations that lead to malignant tumors [191,192]. p53 is the principal responder to various cellular stress signals, such as hypoxia, oxidative stress, oncogenic activation, DNA damage, ribosomal stress, and telomere erosion. The tetrameric transcription factor p53 is activated through multiple phosphorylation events and, depending on the type of stress, this activation results in upregulation or repression of genes involved in cell cycle arrest to restore genetic integrity and DNA repair, apoptosis, senescence, autophagy, or ferroptosis to eliminate unrecoverable cells. p53 also regulates genes involved in anti-angiogenesis, protection against oxidative stress, the regulation of metabolic homeostasis, and stem cell maintenance roles. In unstressed cells, p53 protein levels are regulated via a negative-feedback loop, whereby p53 induces the transcription of its own negative regulator, MDM2, that ubiquitinates p53 and marks it for proteasomal degradation. p53 is inactivated in almost every tumor, through either mutation in the p53 gene or the deregulation of its associated pathways [191,192,193]. The majority of p53 mutations are missense mutations that lead to the synthesis of p53 proteins unable to bind the target gene promoters of wild-type p53 [194]. Mutated p53 can sequester various tumor suppressors, including non-mutated p53 (dominant-negative function) and the family members p63 and p73, inhibiting their pro-apoptotic function [195]. Most p53 missense mutants acquire oncogenic gain-of-function activities that allow them to interact with other transcription factors, including NF-Y, Sp1, ETS1/2, NF-kB, and SMADs [196]. These changes lead to increased genetic instability, cellular proliferation, metastasis, and chemo-/radio-resistance [197].
p53 has been widely involved in the study of neurodegenerative diseases [198,199]. In some of the most common disorders, p53 activity and expression increase in human and mouse model brains [200,201,202,203]. Interestingly, in AD or PD disease, besides high p53 levels, the subcellular location is compromised in the diseased brain [204]. It was observed that whereas p53 and its phosphorylated form (p-p53) are found mainly in the nucleus of control patients, in the brain of a patient with AD, it is located almost exclusively in the cytoplasm, revealing an altered transport cytoplasm-nucleus that might be relevant for the involvement of p53 in the pathological progression of neurological disorders [204]. The formation of p53 aggregates and the destabilization of the microtubules network in the perinuclear area of neurons could be one of the reasons for this cytoplasm-nucleus transport.
Cytoplasmic p53 has been involved in both tau and amyloid pathologies in AD brains. In humans, p53 interacts with Tau and PS1. Several AD mouse models showed that Tau, APP, and PS1 expression levels modulate p53 expression. In this regard, genetic deletions of PS1 and APP decrease the expression of p53 in the mouse brain [198,205]. Increasing evidence showed that p53 is also a repressor of the activity of the different genes involved in the development of neurodegenerative diseases, such as Tau and PS1 in AD [206], and Parkin and α-synuclein in PD. p53 controls the expression of these genes and thus, in turn, can modulate the activity of p53, creating a regulatory loop where higher levels of p53 lead to a repression of PS1, Tau, or Parkin with a concomitant decrease of p53 activity. Alterations in the expression of PS1, Tau, or Parkin led to an imbalance in the regulation of p53 that could have important pathological consequences [199,204,205].
The role of p53 at the onset and development of HD has been described in depth. p53 levels are high in the brains of HD patients, and its expression also positively correlates with the grade of the disorder [207]. It has been found that p53 binds HTT. Mouse models overexpressing mutant forms of HTT.
Present increased levels of p53. In these models, genetic deletions of p53, rescued neurodegeneration showed in HD and the neurodevelopmental abnormalities associated with these models [207,208].
In PD, p53 is an important player in dopaminergic cell loss. Specific ablation of p53 in dopaminergic neurons protects against MPTP-induced neurodegeneration and improves the motor coordination found in this pharmacological approach of PD [209].

2.9. Cell Cycle Pathway

The cell cycle is a complex and orchestrated process that ensures duplication of the genetic material and cell division [210]. This pathway is highly regulated to avoid the transmission of the altered genome to daughter cells. There are checkpoints to regulate the cell cycle, inducing arrests for cell cycle progression and promoting DNA repair or, in case of unrepairable damage, stimulating cell death. The cell cycle consists of four phases: G0/G1, S, G2, and M. The progression is through CDKs and cyclin proteins activation by phosphorylation. In cancer cells, the aberrant activity of the cell cycle is due to mutations in genes encoding cell cycle proteins or components of upstream signaling pathways. For example, CDKN2A (encodes tumor suppressors p16INK4A and p14ARF) and CDKN2B genes (encodes tumor suppressors p16INK4A-p14ARF and p15INK, respectively) are commonly deleted, or its promoter is silenced by methylation in human cancers [210,211]. Around 54% of glioblastomas present deep deletion of cdkn2a or cdkn2b genes. CDK4 and cyclin D1 (CCDN1) locus is frequently amplified in human cancers. CDK4 is amplified in 20% of glioblastomas and sarcomas and CCDN1 is amplified in 32% of esophageal adenocarcinoma [36,171,172].
Despite neurons being postmitotic cells, and hence they are in a quiescent state, dysregulations in their cell cycle have been observed in many of the most common neurodegenerative disorders [212,213]. Neurons of diseased brains have an aberrant DNA replication. These neurons can re-enter into the cell cycle and start their DNA replication but are not able to divide. The absence of mitosis has a detrimental effect on the mature neurons, which may be associated with the development of pathologies concomitant to neurological disorder’s progress [212]. Changes in different cyclins, CDKs, and related genes have been found in postmortem brains of AD, PD, HD, and ALS patients (reviewed in [214]). In different areas of the AD brain, cyclin B, D, CDK4, and CDK5 levels, among others, are upregulated [214,215]. The activators of the CDKs are also highly expressed in AD [214,216]. Moreover, CDK2, CDK5, PCNA, and Rb are expressed aberrantly in the SN of PD patients, and HD brains present increased cyclin D1 levels [217]. The expression of mutant htt leads to neurons re-entering the cell cycle [218].
In mouse models the potential involvement was also confirmed of cell cycle re-entry found in human brains with different diseases. In AD, both pharmacologic and genetic induction of Aβ accumulation in the mouse brain triggers the expression of different genes involved in the cell cycle re-entry. A recent study shows increased expression of an S/G2/M marker in the hippocampus of AD patients and mouse models [213]. Interestingly, authors reported that neurons that undergo rapid cell cycle re-entry showed protective effects against amyloid-induced neuronal death. Dysregulation of the cell cycle in AD has been linked to hyperphosphorylation of Tau. CDK1, CDK2, or cyclin B colocalize with NFTs, and CDK5 expression induces the hyperphosphorylation of Tau. Disruption of the cell cycle has been proposed as a therapeutic target in AD. Genetic activation of the cell cycle in the mouse brain triggers progressive neurodegeneration associated with an increased amyloid load and NFT number [219]. In turn, pharmacological inhibition of specific genes related to the cell cycle alleviates the pathologies of AD mouse models [216].
Similar to AD, SN neurons of mouse models of PD present deregulated cell cycles. MPTP treatment induces the expression and activity of CDK5 in dopaminergic neurons, and its pharmacological inhibition attenuates the MPTP-induced dopamine cell loss [220]. α-synuclein, another hallmark of PD, has been documented to increase the expression of cyclin B [221,222].

2.10. PI3K/AKT/mTOR Pathway

The PI3K/AKT/mTOR (phosphatidylinositol 3-kinase/protein kinase-B/mechanistic target of rapamycin) signaling pathway is one of the most signally altered in cancer [223]. This pathway controls multiple cellular processes, including proliferation, survival, differentiation, metabolism, motility transcription, and protein synthesis [224]. In cancer, this pathway is hyperactivated by different genetic alterations, inducing tumorigenesis, proliferation, apoptosis, metastasis, EMT, stem-like phenotype, immune microenvironment, or drug resistance [225]. Very simplified, RTKs activate PI3K, which are heterodimers consisting of p110 catalytic and p85 regulatory subunits. P110 phosphorylate phosphatidylinositol-4,5-bisphosphate (PIP2) to generate phosphatidylinositol-3,4,5-trisphosphate (PIP3). PIP3 recruits AKT and PDK1. AKT is phosphorylated at Thr308 by PDK1 and at Ser473 by mTOR complex 2 (mTORC2), which increases its kinase activity. AKT phosphorylates TSC2 and TSC1 (tuberous sclerosis proteins 1 and 2) and dissociates the TSC1–TSC2 complex. The TSC1–TSC2 complex negatively regulates the activity of the kinase mTOR; therefore, AKT results in the activation of mTORC1 [226]. PTEN and INPP4B are negative regulators of PI3K activation [227]. The most predominant alterations in the PI3K pathway are activating events (mutations or amplification) in PIK3CA (gene encoding p110) and inactivation events (mutations or loss) in PTEN or PIK3R1. Less common alterations in AKT, TSC1, TSC2, LKB1, mTOR and other critical genes have also been found in cancer.
Dysregulations in the different components of the PI3K pathway have been reported in the development of neurodegenerative diseases [228,229,230]. Although with controversy, signaling activation might happen in AD and PD. Significant activation (phosphorylation) of AKT was observed in neurons of postmortem brains of AD patients. With no reported changes in total levels of AKT, p-AKT, not only was found upregulated [231] but was also located near the nuclear envelope. Other studies reported decreased activation of the PI3K/AKT pathway in AD brains. There is more consensus in the pathway role in PD; AKT activity decreased in the postmortem brains of PD patients [232,233,234,235]. Diverse studies have shown the protective role of overexpressed AKT in PD mouse models [236,237,238].
PI3K/AKT signaling has a myriad of downstream components. Among others, mTOR and GSK3β are two AKT targets involved in the control of autophagy, amyloid aggregation, and Tau phosphorylation in the progression of neurodegenerative disorders, respectively. Activated AKT induces the phosphorylation of both mTOR and GSK3β. Whereas p-mTOR is the active form, phosphorylation represses the activity of GSk3β. PI3K/AKT/mTOR pathway has been linked to disrupted autophagic processes in the human AD brain. At the onset of AD, there were increased mTOR levels [239] and correlated with diminished levels of autophagy markers. In line with these findings, mTOR levels increased during the appearance of pathologies in an AD mouse model.
The PiI3K/AKT/GSK3β pathway has an important role in the hyperphosphorylation of Tau. GS3Kβ has been pinpointed as one of the most important players in Tau phosphorylation, and the consequent accumulation of NFTs reports linking AKT with GS3Kβ might also be confusing [239,240]. Increased levels of Akt found in AD brains correlated with lower activation of GSK3β. Inactivated GSK3β should diminish the levels of pTAU, but rather than this, it is described that pTAU levels are increased in the diseased brain, suggesting the implication of other kinases in the onset and progression of tautologies [239].

3. Concluding Remarks

In this review we describe the individual role and genetic alterations of the significant components of the canonical molecular cancer pathway in the context of progression of different neurodegenerative diseases (Figure 1). Effectors of these signalling pathways play crucial functions in the appearance of the most characteristic pathologies. However, the intense crosstalk between the pathways makes the identification of one individual therapeutical target complicated from a molecular point of view. Despite all the characterization of the different signalling pathways, more efforts are required to understand the global mechanisms shared by the different pathways, with the final goal of obtaining a molecular understanding of the onset and progression of neurodegenerative diseases. This would advance the development of future therapeutical treatments against sporadic degenerative diseases.
Despite this review focusing on the most altered signaling pathways in the different neurodegenerative diseases, other factors are also involved in the appearance of their pathologies. For example, the role of metabolism in the control of cognitive functions has gained relevance in the last few years [241]. It is known that alterations in energy metabolism lead to pathologies associated with neurodegeneration [241]. Peripheral and central immune systems and their crosstalk have been also pointed out as factors involved in age-related diseases [242]. Although their contribution is clear, the specific mechanisms are still not fully understood. The identification of these and other mechanisms, together with the molecular pathways, will lead to finding novel strategies for therapeutical treatments. One of the latest strategies is the implementation of stem cell therapies, whose final goal is to minimize neuronal loss [243].

Author Contributions

Conceptualizing, writing, and editing of the manuscript, L.V. and M.E.R.G.-R. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Ayudas Atracción de Talento, Modalidad 1: 2019T1/BMD13039, Comunidad de Madrid (MERGR).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bray, F.; Laversanne, M.; Weiderpass, E.; Soerjomataram, I. The ever-increasing importance of cancer as a leading cause of premature death worldwide. Cancer 2021, 127, 3029–3030. [Google Scholar] [CrossRef] [PubMed]
  2. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global cancer statistics 2020: Globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  3. Hou, Y.; Dan, X.; Babbar, M.; Wei, Y.; Hasselbalch, S.G.; Croteau, D.L.; Bohr, V.A. Ageing as a risk factor for neurodegenerative disease. Nat. Rev. Neurol. 2019, 15, 565–581. [Google Scholar] [CrossRef] [PubMed]
  4. Global Burden of Disease Collaborators. Global, regional, and national burden of neurological disorders, 1990–2016: A systematic analysis for the global burden of disease study 2016. Lancet Neurol. 2019, 18, 459–480. [Google Scholar] [CrossRef] [Green Version]
  5. Yang, W.; Hamilton, J.L.; Kopil, C.; Beck, J.C.; Tanner, C.M.; Albin, R.L.; Ray Dorsey, E.; Dahodwala, N.; Cintina, I.; Hogan, P.; et al. Current and projected future economic burden of parkinson’s disease in the U.S. NPJ. Parkinsons Dis. 2020, 6, 15. [Google Scholar] [CrossRef] [PubMed]
  6. Dugger, B.N.; Dickson, D.W. Pathology of neurodegenerative diseases. Cold Spring Harb. Perspect. Biol. 2017, 9, a028035. [Google Scholar] [CrossRef] [PubMed]
  7. Bloom, G.S. Amyloid-beta and tau: The trigger and bullet in alzheimer disease pathogenesis. JAMA Neurol. 2014, 71, 505–508. [Google Scholar] [CrossRef] [Green Version]
  8. Antony, P.M.; Diederich, N.J.; Kruger, R.; Balling, R. The hallmarks of parkinson’s disease. FEBS J. 2013, 280, 5981–5993. [Google Scholar] [CrossRef] [Green Version]
  9. Klein, J.A.; Ackerman, S.L. Oxidative stress, cell cycle, and neurodegeneration. J. Clin. Investig. 2003, 111, 785–793. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Gkekas, I.; Gioran, A.; Boziki, M.K.; Grigoriadis, N.; Chondrogianni, N.; Petrakis, S. Oxidative stress and neurodegeneration: Interconnected processes in polyq diseases. Antioxidants 2021, 10, 1450. [Google Scholar] [CrossRef]
  11. Tanzi, R.E. The genetics of alzheimer disease. Cold Spring Harb. Perspect. Med. 2012, 2, a006296. [Google Scholar] [CrossRef] [PubMed]
  12. Aasly, J.O. Long-term outcomes of genetic parkinson’s disease. J. Mov. Disord. 2020, 13, 81–96. [Google Scholar] [CrossRef]
  13. Bates, G.P. History of genetic disease: The molecular genetics of huntington disease—A history. Nat. Rev. Genet. 2005, 6, 766–773. [Google Scholar] [CrossRef] [PubMed]
  14. Frisoni, G.B.; Laakso, M.P.; Beltramello, A.; Geroldi, C.; Bianchetti, A.; Soininen, H.; Trabucchi, M. Hippocampal and entorhinal cortex atrophy in frontotemporal dementia and alzheimer’s disease. Neurology 1999, 52, 91–100. [Google Scholar] [CrossRef]
  15. Olivieri, P.; Lagarde, J.; Lehericy, S.; Valabregue, R.; Michel, A.; Mace, P.; Caille, F.; Gervais, P.; Bottlaender, M.; Sarazin, M. Early alteration of the locus coeruleus in phenotypic variants of alzheimer’s disease. Ann. Clin. Transl. Neurol. 2019, 6, 1345–1351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Mu, Y.; Gage, F.H. Adult hippocampal neurogenesis and its role in alzheimer’s disease. Mol. Neurodegener. 2011, 6, 85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Chen, X.Q.; Mobley, W.C. Exploring the pathogenesis of alzheimer disease in basal forebrain cholinergic neurons: Converging insights from alternative hypotheses. Front. Neurosci. 2019, 13, 446. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Thal, D.R.; Rub, U.; Orantes, M.; Braak, H. Phases of a beta-deposition in the human brain and its relevance for the development of ad. Neurology 2002, 58, 1791–1800. [Google Scholar] [CrossRef]
  19. Van Hoesen, G.W.; Hyman, B.T.; Damasio, A.R. Entorhinal cortex pathology in alzheimer’s disease. Hippocampus 1991, 1, 1–8. [Google Scholar] [CrossRef] [PubMed]
  20. Alexander, G.E. Biology of parkinson’s disease: Pathogenesis and pathophysiology of a multisystem neurodegenerative disorder. Dialogues Clin. Neurosci. 2004, 6, 259–280. [Google Scholar] [PubMed]
  21. Damier, P.; Hirsch, E.C.; Agid, Y.; Graybiel, A.M. The substantia nigra of the human brain. Ii. Patterns of loss of dopamine-containing neurons in parkinson’s disease. Brain 1999, 122, 1437–1448. [Google Scholar] [CrossRef] [PubMed]
  22. Fearnley, J.M.; Lees, A.J. Ageing and parkinson’s disease: Substantia nigra regional selectivity. Brain 1991, 114, 2283–2301. [Google Scholar] [CrossRef]
  23. Alberico, S.L.; Cassell, M.D.; Narayanan, N.S. The vulnerable ventral tegmental area in parkinson’s disease. Basal Ganglia 2015, 5, 51–55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Vonsattel, J.P.; DiFiglia, M. Huntington disease. J. Neuropathol. Exp. Neurol. 1998, 57, 369–384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Tang, T.S.; Chen, X.; Liu, J.; Bezprozvanny, I. Dopaminergic signaling and striatal neurodegeneration in huntington’s disease. J. Neurosci. 2007, 27, 7899–7910. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Kiernan, M.C.; Vucic, S.; Cheah, B.C.; Turner, M.R.; Eisen, A.; Hardiman, O.; Burrell, J.R.; Zoing, M.C. Amyotrophic lateral sclerosis. Lancet 2011, 377, 942–955. [Google Scholar] [CrossRef] [Green Version]
  27. Renton, A.E.; Chio, A.; Traynor, B.J. State of play in amyotrophic lateral sclerosis genetics. Nat. Neurosci. 2014, 17, 17–23. [Google Scholar] [CrossRef] [PubMed]
  28. Taylor, J.P.; Brown, R.H., Jr.; Cleveland, D.W. Decoding als: From genes to mechanism. Nature 2016, 539, 197–206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Bocchetta, M.; Iglesias, J.E.; Cash, D.M.; Warren, J.D.; Rohrer, J.D. Amygdala subnuclei are differentially affected in the different genetic and pathological forms of frontotemporal dementia. Alzheimer Dement. 2019, 11, 136–141. [Google Scholar] [CrossRef] [PubMed]
  30. Dickson, D.W. Pick’s disease: A modern approach. Brain Pathol. 1998, 8, 339–354. [Google Scholar] [CrossRef] [PubMed]
  31. Dickson, D.W. Neuropathology of pick’s disease. Neurology 2001, 56, S16–S20. [Google Scholar] [CrossRef] [PubMed]
  32. Tsuchiya, K.; Ikeda, M.; Hasegawa, K.; Fukui, T.; Kuroiwa, T.; Haga, C.; Oyanagi, S.; Nakano, I.; Matsushita, M.; Yagishita, S.; et al. Distribution of cerebral cortical lesions in pick’s disease with pick bodies: A clinicopathological study of six autopsy cases showing unusual clinical presentations. Acta Neuropathol. 2001, 102, 553–571. [Google Scholar] [CrossRef]
  33. Hanahan, D. Hallmarks of cancer: New dimensions. Cancer Discov. 2022, 12, 31–46. [Google Scholar] [CrossRef]
  34. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Hanahan, D.; Weinberg, R.A. The hallmarks of cancer. Cell 2000, 100, 57–70. [Google Scholar] [CrossRef] [Green Version]
  36. Sanchez-Vega, F.; Mina, M.; Armenia, J.; Chatila, W.K.; Luna, A.; La, K.C.; Dimitriadoy, S.; Liu, D.L.; Kantheti, H.S.; Saghafinia, S.; et al. Oncogenic signaling pathways in the cancer genome atlas. Cell 2018, 173, 321–337.e10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Zheng, Y.; Pan, D. The hippo signaling pathway in development and disease. Dev. Cell 2019, 50, 264–282. [Google Scholar] [CrossRef] [PubMed]
  38. Halder, G.; Johnson, R.L. Hippo signaling: Growth control and beyond. Development 2011, 138, 9–22. [Google Scholar] [CrossRef] [Green Version]
  39. Zhang, L. Control of growth and beyond: A special issue on hippo signaling. Acta Biochim. Biophys. Sin 2015, 47, 1. [Google Scholar] [CrossRef] [Green Version]
  40. Harvey, K.F.; Zhang, X.; Thomas, D.M. The hippo pathway and human cancer. Nat. Rev. Cancer 2013, 13, 246–257. [Google Scholar] [CrossRef]
  41. Liu-Chittenden, Y.; Huang, B.; Shim, J.S.; Chen, Q.; Lee, S.J.; Anders, R.A.; Liu, J.O.; Pan, D. Genetic and pharmacological disruption of the tead-yap complex suppresses the oncogenic activity of yap. Genes Dev. 2012, 26, 1300–1305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Zanconato, F.; Cordenonsi, M.; Piccolo, S. Yap/taz at the roots of cancer. Cancer Cell 2016, 29, 783–803. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Yu, F.X.; Zhang, Y.; Park, H.W.; Jewell, J.L.; Chen, Q.; Deng, Y.; Pan, D.; Taylor, S.S.; Lai, Z.C.; Guan, K.L. Protein kinase a activates the hippo pathway to modulate cell proliferation and differentiation. Genes Dev. 2013, 27, 1223–1232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Zhao, B.; Wei, X.; Li, W.; Udan, R.S.; Yang, Q.; Kim, J.; Xie, J.; Ikenoue, T.; Yu, J.; Li, L.; et al. Inactivation of yap oncoprotein by the hippo pathway is involved in cell contact inhibition and tissue growth control. Genes Dev. 2007, 21, 2747–2761. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. James, M.F.; Han, S.; Polizzano, C.; Plotkin, S.R.; Manning, B.D.; Stemmer-Rachamimov, A.O.; Gusella, J.F.; Ramesh, V. Nf2/merlin is a novel negative regulator of mtor complex 1, and activation of mtorc1 is associated with meningioma and schwannoma growth. Mol. Cell Biol. 2009, 29, 4250–4261. [Google Scholar] [CrossRef] [Green Version]
  46. Lopez-Lago, M.A.; Okada, T.; Murillo, M.M.; Socci, N.; Giancotti, F.G. Loss of the tumor suppressor gene nf2, encoding merlin, constitutively activates integrin-dependent mtorc1 signaling. Mol. Cell Biol. 2009, 29, 4235–4249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Lin, L.; Sabnis, A.J.; Chan, E.; Olivas, V.; Cade, L.; Pazarentzos, E.; Asthana, S.; Neel, D.; Yan, J.J.; Lu, X.; et al. The hippo effector yap promotes resistance to raf- and mek-targeted cancer therapies. Nat. Genet. 2015, 47, 250–256. [Google Scholar] [CrossRef]
  48. Kurppa, K.J.; Liu, Y.; To, C.; Zhang, T.; Fan, M.; Vajdi, A.; Knelson, E.H.; Xie, Y.; Lim, K.; Cejas, P.; et al. Treatment-induced tumor dormancy through yap-mediated transcriptional reprogramming of the apoptotic pathway. Cancer Cell 2020, 37, 104–122.e112. [Google Scholar] [CrossRef] [PubMed]
  49. Sahu, M.R.; Mondal, A.C. Neuronal hippo signaling: From development to diseases. Dev. Neurobiol. 2021, 81, 92–109. [Google Scholar] [CrossRef]
  50. Sahu, M.R.; Mondal, A.C. The emerging role of hippo signaling in neurodegeneration. J. Neurosci. Res. 2020, 98, 796–814. [Google Scholar] [CrossRef]
  51. Bruno, L.; Karagil, S.; Mahmood, A.; Elbediwy, A.; Stolinski, M.; Mackenzie, F.E. Mechanosensing and the hippo pathway in microglia: A potential link to alzheimer’s disease pathogenesis? Cells 2021, 10, 3144. [Google Scholar] [CrossRef] [PubMed]
  52. Xu, J.; Patassini, S.; Rustogi, N.; Riba-Garcia, I.; Hale, B.D.; Phillips, A.M.; Waldvogel, H.; Haines, R.; Bradbury, P.; Stevens, A.; et al. Regional protein expression in human alzheimer’s brain correlates with disease severity. Commun. Biol. 2019, 2, 43. [Google Scholar] [CrossRef] [PubMed]
  53. Yamanishi, E.; Hasegawa, K.; Fujita, K.; Ichinose, S.; Yagishita, S.; Murata, M.; Tagawa, K.; Akashi, T.; Eishi, Y.; Okazawa, H. A novel form of necrosis, triad, occurs in human huntington’s disease. Acta Neuropathol. Commun. 2017, 5, 19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Tanaka, H.; Homma, H.; Fujita, K.; Kondo, K.; Yamada, S.; Jin, X.; Waragai, M.; Ohtomo, G.; Iwata, A.; Tagawa, K.; et al. Yap-dependent necrosis occurs in early stages of alzheimer’s disease and regulates mouse model pathology. Nat. Commun. 2020, 11, 507. [Google Scholar] [CrossRef] [PubMed]
  55. Mueller, K.A.; Glajch, K.E.; Huizenga, M.N.; Wilson, R.A.; Granucci, E.J.; Dios, A.M.; Tousley, A.R.; Iuliano, M.; Weisman, E.; LaQuaglia, M.J.; et al. Hippo signaling pathway dysregulation in human huntington’s disease brain and neuronal stem cells. Sci. Rep. 2018, 8, 11355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Mao, Y.; Chen, X.; Xu, M.; Fujita, K.; Motoki, K.; Sasabe, T.; Homma, H.; Murata, M.; Tagawa, K.; Tamura, T.; et al. Targeting tead/yap-transcription-dependent necrosis, triad, ameliorates huntington’s disease pathology. Hum. Mol. Genet. 2016, 25, 4749–4770. [Google Scholar] [CrossRef] [PubMed]
  57. Ahn, E.H.; Kang, S.S.; Qi, Q.; Liu, X.; Ye, K. Netrin1 deficiency activates mst1 via unc5b receptor, promoting dopaminergic apoptosis in parkinson’s disease. Proc. Natl. Acad. Sci. USA 2020, 117, 24503–24513. [Google Scholar] [CrossRef] [PubMed]
  58. Lee, J.K.; Shin, J.H.; Hwang, S.G.; Gwag, B.J.; McKee, A.C.; Lee, J.; Kowall, N.W.; Ryu, H.; Lim, D.S.; Choi, E.J. Mst1 functions as a key modulator of neurodegeneration in a mouse model of als. Proc. Natl. Acad. Sci. USA 2013, 110, 12066–12071. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Kovall, R.A.; Gebelein, B.; Sprinzak, D.; Kopan, R. The canonical notch signaling pathway: Structural and biochemical insights into shape, sugar, and force. Dev. Cell 2017, 41, 228–241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Sjoqvist, M.; Andersson, E.R. Do as I say, not(ch) as I do: Lateral control of cell fate. Dev. Biol. 2019, 447, 58–70. [Google Scholar] [CrossRef]
  61. Bray, S.J. Notch signalling in context. Nat. Rev. Mol. Cell Biol. 2016, 17, 722–735. [Google Scholar] [CrossRef] [PubMed]
  62. Kopan, R.; Ilagan, M.X. The canonical notch signaling pathway: Unfolding the activation mechanism. Cell 2009, 137, 216–233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Schwanbeck, R.; Martini, S.; Bernoth, K.; Just, U. The notch signaling pathway: Molecular basis of cell context dependency. Eur. J. Cell Biol. 2011, 90, 572–581. [Google Scholar] [CrossRef] [PubMed]
  64. Reedijk, M.; Odorcic, S.; Chang, L.; Zhang, H.; Miller, N.; McCready, D.R.; Lockwood, G.; Egan, S.E. High-level coexpression of jag1 and notch1 is observed in human breast cancer and is associated with poor overall survival. Cancer Res. 2005, 65, 8530–8537. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Ranganathan, P.; Weaver, K.L.; Capobianco, A.J. Notch signalling in solid tumours: A little bit of everything but not all the time. Nat. Rev. Cancer 2011, 11, 338–351. [Google Scholar] [CrossRef] [PubMed]
  66. Ferrarotto, R.; Mitani, Y.; Diao, L.; Guijarro, I.; Wang, J.; Zweidler-McKay, P.; Bell, D.; William, W.N., Jr.; Glisson, B.S.; Wick, M.J.; et al. Activating notch1 mutations define a distinct subgroup of patients with adenoid cystic carcinoma who have poor prognosis, propensity to bone and liver metastasis, and potential responsiveness to notch1 inhibitors. J. Clin. Oncol. 2017, 35, 352–360. [Google Scholar] [CrossRef]
  67. Tosello, V.; Ferrando, A.A. The notch signaling pathway: Role in the pathogenesis of t-cell acute lymphoblastic leukemia and implication for therapy. Ther. Adv. Hematol. 2013, 4, 199–210. [Google Scholar] [CrossRef]
  68. Woo, H.N.; Park, J.S.; Gwon, A.R.; Arumugam, T.V.; Jo, D.G. Alzheimer’s disease and notch signaling. Biochem. Biophys. Res. Commun. 2009, 390, 1093–1097. [Google Scholar] [CrossRef]
  69. Basak, O.; Giachino, C.; Fiorini, E.; Macdonald, H.R.; Taylor, V. Neurogenic subventricular zone stem/progenitor cells are notch1-dependent in their active but not quiescent state. J. Neurosci. 2012, 32, 5654–5666. [Google Scholar] [CrossRef] [Green Version]
  70. Cho, S.J.; Yun, S.M.; Jo, C.; Jeong, J.; Park, M.H.; Han, C.; Koh, Y.H. Altered expression of notch1 in alzheimer’s disease. PLoS ONE 2019, 14, e0224941. [Google Scholar] [CrossRef]
  71. Moehlmann, T.; Winkler, E.; Xia, X.; Edbauer, D.; Murrell, J.; Capell, A.; Kaether, C.; Zheng, H.; Ghetti, B.; Haass, C.; et al. Presenilin-1 mutations of leucine 166 equally affect the generation of the notch and app intracellular domains independent of their effect on abeta 42 production. Proc. Natl. Acad. Sci. USA 2002, 99, 8025–8030. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Chavez-Gutierrez, L.; Bammens, L.; Benilova, I.; Vandersteen, A.; Benurwar, M.; Borgers, M.; Lismont, S.; Zhou, L.; Van Cleynenbreugel, S.; Esselmann, H.; et al. The mechanism of gamma-secretase dysfunction in familial alzheimer disease. EMBO J. 2012, 31, 2261–2274. [Google Scholar] [CrossRef] [PubMed]
  73. Berezovska, O.; Xia, M.Q.; Hyman, B.T. Notch is expressed in adult brain, is coexpressed with presenilin-1, and is altered in alzheimer disease. J. Neuropathol. Exp. Neurol. 1998, 57, 738–745. [Google Scholar] [CrossRef] [Green Version]
  74. Nagarsheth, M.H.; Viehman, A.; Lippa, S.M.; Lippa, C.F. Notch-1 immunoexpression is increased in alzheimer’s and pick’s disease. J. Neurol. Sci. 2006, 244, 111–116. [Google Scholar] [CrossRef]
  75. Brai, E.; Alina Raio, N.; Alberi, L. Notch1 hallmarks fibrillary depositions in sporadic alzheimer’s disease. Acta Neuropathol. Commun. 2016, 4, 64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Marathe, S.; Jaquet, M.; Annoni, J.M.; Alberi, L. Jagged1 is altered in alzheimer’s disease and regulates spatial memory processing. Front. Cell Neurosci. 2017, 11, 220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Imai, Y.; Kobayashi, Y.; Inoshita, T.; Meng, H.; Arano, T.; Uemura, K.; Asano, T.; Yoshimi, K.; Zhang, C.L.; Matsumoto, G.; et al. The parkinson’s disease-associated protein kinase lrrk2 modulates notch signaling through the endosomal pathway. PLoS Genet. 2015, 11, e1005503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Fischer, D.F.; van Dijk, R.; Sluijs, J.A.; Nair, S.M.; Racchi, M.; Levelt, C.N.; van Leeuwen, F.W.; Hol, E.M. Activation of the notch pathway in down syndrome: Cross-talk of notch and app. FASEB J. 2005, 19, 1451–1458. [Google Scholar]
  79. Ma, Q. Role of nrf2 in oxidative stress and toxicity. Annu. Rev. Pharmacol. Toxicol. 2013, 53, 401–426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Zhao, J.; Lin, X.; Meng, D.; Zeng, L.; Zhuang, R.; Huang, S.; Lv, W.; Hu, J. Nrf2 mediates metabolic reprogramming in non-small cell lung cancer. Front. Oncol. 2020, 10, 578315. [Google Scholar] [CrossRef]
  81. Itoh, K.; Wakabayashi, N.; Katoh, Y.; Ishii, T.; Igarashi, K.; Engel, J.D.; Yamamoto, M. Keap1 represses nuclear activation of antioxidant responsive elements by nrf2 through binding to the amino-terminal neh2 domain. Genes Dev. 1999, 13, 76–86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Sporn, M.B.; Liby, K.T. Nrf2 and cancer: The good, the bad and the importance of context. Nat. Rev. Cancer 2012, 12, 564–571. [Google Scholar] [CrossRef] [PubMed]
  83. Baird, L.; Lleres, D.; Swift, S.; Dinkova-Kostova, A.T. Regulatory flexibility in the nrf2-mediated stress response is conferred by conformational cycling of the keap1-nrf2 protein complex. Proc. Natl. Acad. Sci. USA 2013, 110, 15259–15264. [Google Scholar] [CrossRef] [Green Version]
  84. Kitamura, H.; Motohashi, H. Nrf2 addiction in cancer cells. Cancer Sci. 2018, 109, 900–911. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Satoh, H.; Moriguchi, T.; Takai, J.; Ebina, M.; Yamamoto, M. Nrf2 prevents initiation but accelerates progression through the kras signaling pathway during lung carcinogenesis. Cancer Res. 2013, 73, 4158–4168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Tao, S.; Rojo de la Vega, M.; Chapman, E.; Ooi, A.; Zhang, D.D. The effects of nrf2 modulation on the initiation and progression of chemically and genetically induced lung cancer. Mol. Carcinog. 2018, 57, 182–192. [Google Scholar] [CrossRef] [PubMed]
  87. Zgorzynska, E.; Dziedzic, B.; Walczewska, A. An overview of the nrf2/are pathway and its role in neurodegenerative diseases. Int. J. Mol. Sci. 2021, 22, 9592. [Google Scholar] [CrossRef]
  88. Zhang, W.; Feng, C.; Jiang, H. Novel target for treating alzheimer’s diseases: Crosstalk between the nrf2 pathway and autophagy. Ageing Res. Rev. 2021, 65, 101207. [Google Scholar] [CrossRef]
  89. Saha, S.; Buttari, B.; Profumo, E.; Tucci, P.; Saso, L. A perspective on nrf2 signaling pathway for neuroinflammation: A potential therapeutic target in alzheimer’s and parkinson’s diseases. Front. Cell Neurosci. 2021, 15, 787258. [Google Scholar] [CrossRef]
  90. Kersten, S.; Mandard, S.; Tan, N.S.; Escher, P.; Metzger, D.; Chambon, P.; Gonzalez, F.J.; Desvergne, B.; Wahli, W. Characterization of the fasting-induced adipose factor fiaf, a novel peroxisome proliferator-activated receptor target gene. J. Biol. Chem. 2000, 275, 28488–28493. [Google Scholar] [CrossRef] [Green Version]
  91. Delaidelli, A.; Richner, M.; Jiang, L.; van der Laan, A.; Bergholdt Jul Christiansen, I.; Ferreira, N.; Nyengaard, J.R.; Vaegter, C.B.; Jensen, P.H.; Mackenzie, I.R.; et al. Alpha-synuclein pathology in parkinson disease activates homeostatic nrf2 anti-oxidant response. Acta Neuropathol. Commun. 2021, 9, 105. [Google Scholar] [CrossRef] [PubMed]
  92. Ramsey, C.P.; Glass, C.A.; Montgomery, M.B.; Lindl, K.A.; Ritson, G.P.; Chia, L.A.; Hamilton, R.L.; Chu, C.T.; Jordan-Sciutto, K.L. Expression of nrf2 in neurodegenerative diseases. J. Neuropathol. Exp. Neurol. 2007, 66, 75–85. [Google Scholar] [CrossRef] [PubMed]
  93. Ren, P.; Chen, J.; Li, B.; Zhang, M.; Yang, B.; Guo, X.; Chen, Z.; Cheng, H.; Wang, P.; Wang, S.; et al. Nrf2 ablation promotes alzheimer’s disease-like pathology in app/ps1 transgenic mice: The role of neuroinflammation and oxidative stress. Oxid Med. Cell Longev. 2020, 2020, 3050971. [Google Scholar] [CrossRef]
  94. Kanninen, K.; Malm, T.M.; Jyrkkanen, H.K.; Goldsteins, G.; Keksa-Goldsteine, V.; Tanila, H.; Yamamoto, M.; Yla-Herttuala, S.; Levonen, A.L.; Koistinaho, J. Nuclear factor erythroid 2-related factor 2 protects against beta amyloid. Mol. Cell Neurosci. 2008, 39, 302–313. [Google Scholar] [CrossRef] [PubMed]
  95. Kanninen, K.; Heikkinen, R.; Malm, T.; Rolova, T.; Kuhmonen, S.; Leinonen, H.; Yla-Herttuala, S.; Tanila, H.; Levonen, A.L.; Koistinaho, M.; et al. Intrahippocampal injection of a lentiviral vector expressing nrf2 improves spatial learning in a mouse model of alzheimer’s disease. Proc. Natl. Acad. Sci. USA 2009, 106, 16505–16510. [Google Scholar] [CrossRef] [Green Version]
  96. Pajares, M.; Jimenez-Moreno, N.; Garcia-Yague, A.J.; Escoll, M.; de Ceballos, M.L.; Van Leuven, F.; Rabano, A.; Yamamoto, M.; Rojo, A.I.; Cuadrado, A. Transcription factor nfe2l2/nrf2 is a regulator of macroautophagy genes. Autophagy 2016, 12, 1902–1916. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Tanji, K.; Maruyama, A.; Odagiri, S.; Mori, F.; Itoh, K.; Kakita, A.; Takahashi, H.; Wakabayashi, K. Keap1 is localized in neuronal and glial cytoplasmic inclusions in various neurodegenerative diseases. J. Neuropathol. Exp. Neurol. 2013, 72, 18–28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Clevers, H.; Nusse, R. Wnt/beta-catenin signaling and disease. Cell 2012, 149, 1192–1205. [Google Scholar] [CrossRef] [Green Version]
  99. Nusse, R.; Clevers, H. Wnt/beta-catenin signaling, disease, and emerging therapeutic modalities. Cell 2017, 169, 985–999. [Google Scholar] [CrossRef] [PubMed]
  100. Pai, S.G.; Carneiro, B.A.; Mota, J.M.; Costa, R.; Leite, C.A.; Barroso-Sousa, R.; Kaplan, J.B.; Chae, Y.K.; Giles, F.J. Wnt/beta-catenin pathway: Modulating anticancer immune response. J. Hematol. Oncol. 2017, 10, 101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. He, T.C.; Sparks, A.B.; Rago, C.; Hermeking, H.; Zawel, L.; da Costa, L.T.; Morin, P.J.; Vogelstein, B.; Kinzler, K.W. Identification of c-myc as a target of the apc pathway. Science 1998, 281, 1509–1512. [Google Scholar] [CrossRef]
  102. Shtutman, M.; Zhurinsky, J.; Simcha, I.; Albanese, C.; D’Amico, M.; Pestell, R.; Ben-Ze’ev, A. The cyclin d1 gene is a target of the beta-catenin/lef-1 pathway. Proc. Natl. Acad. Sci. USA 1999, 96, 5522–5527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Gan, X.Q.; Wang, J.Y.; Xi, Y.; Wu, Z.L.; Li, Y.P.; Li, L. Nuclear dvl, c-jun, beta-catenin, and tcf form a complex leading to stabilization of beta-catenin-tcf interaction. J. Cell Biol. 2008, 180, 1087–1100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Kristensen, B.W.; Priesterbach-Ackley, L.P.; Petersen, J.K.; Wesseling, P. Molecular pathology of tumors of the central nervous system. Ann. Oncol. 2019, 30, 1265–1278. [Google Scholar] [CrossRef] [PubMed]
  105. Lee, Y.; Lee, J.K.; Ahn, S.H.; Lee, J.; Nam, D.H. WNT signaling in glioblastoma and therapeutic opportunities. Lab. Investig. 2016, 96, 137–150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Zurawel, R.H.; Chiappa, S.A.; Allen, C.; Raffel, C. Sporadic medulloblastomas contain oncogenic beta-catenin mutations. Cancer Res. 1998, 58, 896–899. [Google Scholar]
  107. Silva, R.; Marie, S.K.; Uno, M.; Matushita, H.; Wakamatsu, A.; Rosemberg, S.; Oba-Shinjo, S.M. Ctnnb1, axin1 and apc expression analysis of different medulloblastoma variants. Clinics 2013, 68, 167–172. [Google Scholar] [CrossRef]
  108. Inestrosa, N.C.; Varela-Nallar, L. Wnt signaling in the nervous system and in alzheimer’s disease. J. Mol. Cell Biol. 2014, 6, 64–74. [Google Scholar] [CrossRef]
  109. Palomer, E.; Buechler, J.; Salinas, P.C. Wnt signaling deregulation in the aging and alzheimer’s brain. Front. Cell Neurosci. 2019, 13, 227. [Google Scholar] [CrossRef]
  110. Liu, C.C.; Tsai, C.W.; Deak, F.; Rogers, J.; Penuliar, M.; Sung, Y.M.; Maher, J.N.; Fu, Y.; Li, X.; Xu, H.; et al. Deficiency in lrp6-mediated wnt signaling contributes to synaptic abnormalities and amyloid pathology in alzheimer’s disease. Neuron 2014, 84, 63–77. [Google Scholar] [CrossRef] [Green Version]
  111. Folke, J.; Pakkenberg, B.; Brudek, T. Impaired wnt signaling in the prefrontal cortex of alzheimer’s disease. Mol. Neurobiol. 2019, 56, 873–891. [Google Scholar] [CrossRef] [PubMed]
  112. Caricasole, A.; Copani, A.; Caraci, F.; Aronica, E.; Rozemuller, A.J.; Caruso, A.; Storto, M.; Gaviraghi, G.; Terstappen, G.C.; Nicoletti, F. Induction of dickkopf-1, a negative modulator of the wnt pathway, is associated with neuronal degeneration in alzheimer’s brain. J. Neurosci. 2004, 24, 6021–6027. [Google Scholar] [CrossRef]
  113. Rosi, M.C.; Luccarini, I.; Grossi, C.; Fiorentini, A.; Spillantini, M.G.; Prisco, A.; Scali, C.; Gianfriddo, M.; Caricasole, A.; Terstappen, G.C.; et al. Increased dickkopf-1 expression in transgenic mouse models of neurodegenerative disease. J. Neurochem. 2010, 112, 1539–1551. [Google Scholar] [CrossRef] [PubMed]
  114. Zhang, L.; Deng, J.; Pan, Q.; Zhan, Y.; Fan, J.B.; Zhang, K.; Zhang, Z. Targeted methylation sequencing reveals dysregulated wnt signaling in parkinson disease. J. Genet. Genom. 2016, 43, 587–592. [Google Scholar] [CrossRef] [PubMed]
  115. Hata, A.; Chen, Y.G. Tgf-beta signaling from receptors to smads. Cold Spring Harb. Perspect. Biol. 2016, 8. [Google Scholar] [CrossRef] [Green Version]
  116. Massague, J. How cells read tgf-beta signals. Nat. Rev. Mol. Cell Biol. 2000, 1, 169–178. [Google Scholar] [CrossRef]
  117. Lee, M.K.; Pardoux, C.; Hall, M.C.; Lee, P.S.; Warburton, D.; Qing, J.; Smith, S.M.; Derynck, R. Tgf-beta activates erk map kinase signalling through direct phosphorylation of shca. EMBO J. 2007, 26, 3957–3967. [Google Scholar] [CrossRef] [Green Version]
  118. Sorrentino, A.; Thakur, N.; Grimsby, S.; Marcusson, A.; von Bulow, V.; Schuster, N.; Zhang, S.; Heldin, C.H.; Landstrom, M. The type i tgf-beta receptor engages traf6 to activate tak1 in a receptor kinase-independent manner. Nat. Cell Biol. 2008, 10, 1199–1207. [Google Scholar] [CrossRef]
  119. Principe, D.R.; Diaz, A.M.; Torres, C.; Mangan, R.J.; DeCant, B.; McKinney, R.; Tsao, M.S.; Lowy, A.; Munshi, H.G.; Jung, B.; et al. Tgfbeta engages mek/erk to differentially regulate benign and malignant pancreas cell function. Oncogene 2017, 36, 4336–4348. [Google Scholar] [CrossRef] [Green Version]
  120. David, C.J.; Massague, J. Contextual determinants of tgfbeta action in development, immunity and cancer. Nat. Rev. Mol. Cell Biol. 2018, 19, 419–435. [Google Scholar] [CrossRef]
  121. Ikushima, H.; Miyazono, K. Tgfbeta signalling: A complex web in cancer progression. Nat. Rev. Cancer 2010, 10, 415–424. [Google Scholar] [CrossRef] [PubMed]
  122. Tang, B.; Vu, M.; Booker, T.; Santner, S.J.; Miller, F.R.; Anver, M.R.; Wakefield, L.M. Tgf-beta switches from tumor suppressor to prometastatic factor in a model of breast cancer progression. J. Clin. Investig. 2003, 112, 1116–1124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Wakefield, L.M.; Roberts, A.B. Tgf-beta signaling: Positive and negative effects on tumorigenesis. Curr. Opin. Genet. Dev. 2002, 12, 22–29. [Google Scholar] [CrossRef]
  124. Calon, A.; Lonardo, E.; Berenguer-Llergo, A.; Espinet, E.; Hernando-Momblona, X.; Iglesias, M.; Sevillano, M.; Palomo-Ponce, S.; Tauriello, D.V.; Byrom, D.; et al. Stromal gene expression defines poor-prognosis subtypes in colorectal cancer. Nat. Genet. 2015, 47, 320–329. [Google Scholar] [CrossRef] [Green Version]
  125. Okada, Y.; Wang, T.; Kasai, K.; Suzuki, K.; Takikawa, Y. Regulation of transforming growth factor is involved in the efficacy of combined 5-fluorouracil and interferon alpha-2b therapy of advanced hepatocellular carcinoma. Cell Death Discov. 2018, 4, 42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Tauriello, D.V.F. From poor prognosis to promising treatment. Science 2019, 363, 1051. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Tauriello, D.V.F.; Palomo-Ponce, S.; Stork, D.; Berenguer-Llergo, A.; Badia-Ramentol, J.; Iglesias, M.; Sevillano, M.; Ibiza, S.; Canellas, A.; Hernando-Momblona, X.; et al. Tgfbeta drives immune evasion in genetically reconstituted colon cancer metastasis. Nature 2018, 554, 538–543. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Luckett, K.A.; Cracchiolo, J.R.; Krishnamoorthy, G.P.; Leandro-Garcia, L.J.; Nagarajah, J.; Saqcena, M.; Lester, R.; Im, S.Y.; Zhao, Z.; Lowe, S.W.; et al. Co-inhibition of smad and mapk signaling enhances 124i uptake in braf-mutant thyroid cancers. Endocr. Relat. Cancer 2021, 28, 391–402. [Google Scholar] [CrossRef]
  129. Guo, B.; Wu, S.; Zhu, X.; Zhang, L.; Deng, J.; Li, F.; Wang, Y.; Zhang, S.; Wu, R.; Lu, J.; et al. Micropeptide cip2a-bp encoded by linc00665 inhibits triple-negative breast cancer progression. EMBO J. 2020, 39, e102190. [Google Scholar] [CrossRef]
  130. Kashima, R.; Hata, A. The role of tgf-beta superfamily signaling in neurological disorders. Acta Biochim. Biophys. Sin 2018, 50, 106–120. [Google Scholar] [CrossRef] [Green Version]
  131. Tesseur, I.; Zou, K.; Esposito, L.; Bard, F.; Berber, E.; Can, J.V.; Lin, A.H.; Crews, L.; Tremblay, P.; Mathews, P.; et al. Deficiency in neuronal tgf-beta signaling promotes neurodegeneration and alzheimer’s pathology. J. Clin. Investig. 2006, 116, 3060–3069. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. von Bernhardi, R.; Cornejo, F.; Parada, G.E.; Eugenin, J. Role of tgfbeta signaling in the pathogenesis of alzheimer’s disease. Front. Cell Neurosci. 2015, 9, 426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Mocali, A.; Cedrola, S.; Della Malva, N.; Bontempelli, M.; Mitidieri, V.A.; Bavazzano, A.; Comolli, R.; Paoletti, F.; La Porta, C.A. Increased plasma levels of soluble cd40, together with the decrease of tgf beta 1, as possible differential markers of alzheimer disease. Exp. Gerontol. 2004, 39, 1555–1561. [Google Scholar] [CrossRef]
  134. Juraskova, B.; Andrys, C.; Holmerova, I.; Solichova, D.; Hrnciarikova, D.; Vankova, H.; Vasatko, T.; Krejsek, J. Transforming growth factor beta and soluble endoglin in the healthy senior and in alzheimer’s disease patients. J. Nutr. Health Aging 2010, 14, 758–761. [Google Scholar] [CrossRef] [PubMed]
  135. Burton, T.; Liang, B.; Dibrov, A.; Amara, F. Transforming growth factor-beta-induced transcription of the alzheimer beta-amyloid precursor protein gene involves interaction between the ctcf-complex and smads. Biochem. Biophys. Res. Commun. 2002, 295, 713–723. [Google Scholar] [CrossRef]
  136. Colangelo, V.; Schurr, J.; Ball, M.J.; Pelaez, R.P.; Bazan, N.G.; Lukiw, W.J. Gene expression profiling of 12633 genes in alzheimer hippocampal ca1: Transcription and neurotrophic factor down-regulation and up-regulation of apoptotic and pro-inflammatory signaling. J. Neurosci. Res. 2002, 70, 462–473. [Google Scholar] [CrossRef]
  137. Chalmers, K.A.; Love, S. Phosphorylated smad 2/3 colocalizes with phospho-tau inclusions in pick disease, progressive supranuclear palsy, and corticobasal degeneration but not with alpha-synuclein inclusions in multiple system atrophy or dementia with lewy bodies. J. Neuropathol. Exp. Neurol. 2007, 66, 1019–1026. [Google Scholar] [CrossRef] [Green Version]
  138. Chang, K.H.; Wu, Y.R.; Chen, Y.C.; Chen, C.M. Plasma inflammatory biomarkers for huntington’s disease patients and mouse model. Brain Behav. Immun. 2015, 44, 121–127. [Google Scholar] [CrossRef]
  139. Chalmers, K.A.; Love, S. Neurofibrillary tangles may interfere with smad 2/3 signaling in neurons. J. Neuropathol. Exp. Neurol. 2007, 66, 158–167. [Google Scholar] [CrossRef] [Green Version]
  140. Chen, J.H.; Ke, K.F.; Lu, J.H.; Qiu, Y.H.; Peng, Y.P. Protection of tgf-beta1 against neuroinflammation and neurodegeneration in abeta1-42-induced alzheimer’s disease model rats. PLoS ONE 2015, 10, e0116549. [Google Scholar]
  141. Wyss-Coray, T.; Feng, L.; Masliah, E.; Ruppe, M.D.; Lee, H.S.; Toggas, S.M.; Rockenstein, E.M.; Mucke, L. Increased central nervous system production of extracellular matrix components and development of hydrocephalus in transgenic mice overexpressing transforming growth factor-beta 1. Am. J. Pathol. 1995, 147, 53–67. [Google Scholar]
  142. Lifshitz, V.; Weiss, R.; Levy, H.; Frenkel, D. Scavenger receptor a deficiency accelerates cerebrovascular amyloidosis in an animal model. J. Mol. Neurosci. 2013, 50, 198–203. [Google Scholar] [CrossRef] [PubMed]
  143. Vawter, M.P.; Dillon-Carter, O.; Tourtellotte, W.W.; Carvey, P.; Freed, W.J. Tgfbeta1 and tgfbeta2 concentrations are elevated in parkinson’s disease in ventricular cerebrospinal fluid. Exp. Neurol. 1996, 142, 313–322. [Google Scholar] [CrossRef] [PubMed]
  144. Tesseur, I.; Nguyen, A.; Chang, B.; Li, L.; Woodling, N.S.; Wyss-Coray, T.; Luo, J. Deficiency in neuronal tgf-beta signaling leads to nigrostriatal degeneration and activation of tgf-beta signaling protects against mptp neurotoxicity in mice. J. Neurosci. 2017, 37, 4584–4592. [Google Scholar] [CrossRef] [PubMed]
  145. Kandasamy, M.; Aigner, L. Reactive neuroblastosis in huntington’s disease: A putative therapeutic target for striatal regeneration in the adult brain. Front. Cell Neurosci. 2018, 12, 37. [Google Scholar] [CrossRef] [PubMed]
  146. Malynn, B.A.; de Alboran, I.M.; O’Hagan, R.C.; Bronson, R.; Davidson, L.; DePinho, R.A.; Alt, F.W. N-myc can functionally replace c-myc in murine development, cellular growth, and differentiation. Genes Dev. 2000, 14, 1390–1399. [Google Scholar] [CrossRef] [PubMed]
  147. Blackwell, T.K.; Huang, J.; Ma, A.; Kretzner, L.; Alt, F.W.; Eisenman, R.N.; Weintraub, H. Binding of myc proteins to canonical and noncanonical DNA sequences. Mol. Cell Biol. 1993, 13, 5216–5224. [Google Scholar]
  148. Fernandez, P.C.; Frank, S.R.; Wang, L.; Schroeder, M.; Liu, S.; Greene, J.; Cocito, A.; Amati, B. Genomic targets of the human c-myc protein. Genes Dev. 2003, 17, 1115–1129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Hsieh, A.L.; Dang, C.V. Myc, metabolic synthetic lethality, and cancer. Recent Results Cancer Res. 2016, 207, 73–91. [Google Scholar] [PubMed]
  150. Kress, T.R.; Sabo, A.; Amati, B. Myc: Connecting selective transcriptional control to global rna production. Nat. Rev. Cancer 2015, 15, 593–607. [Google Scholar] [CrossRef]
  151. Chang, T.C.; Zeitels, L.R.; Hwang, H.W.; Chivukula, R.R.; Wentzel, E.A.; Dews, M.; Jung, J.; Gao, P.; Dang, C.V.; Beer, M.A.; et al. Lin-28b transactivation is necessary for myc-mediated let-7 repression and proliferation. Proc. Natl. Acad. Sci. USA 2009, 106, 3384–3389. [Google Scholar] [CrossRef] [Green Version]
  152. Ji, H.; Wu, G.; Zhan, X.; Nolan, A.; Koh, C.; De Marzo, A.; Doan, H.M.; Fan, J.; Cheadle, C.; Fallahi, M.; et al. Cell-type independent myc target genes reveal a primordial signature involved in biomass accumulation. PLoS ONE 2011, 6, e26057. [Google Scholar] [CrossRef] [PubMed]
  153. Koh, C.M.; Gurel, B.; Sutcliffe, S.; Aryee, M.J.; Schultz, D.; Iwata, T.; Uemura, M.; Zeller, K.I.; Anele, U.; Zheng, Q.; et al. Alterations in nucleolar structure and gene expression programs in prostatic neoplasia are driven by the myc oncogene. Am. J. Pathol. 2011, 178, 1824–1834. [Google Scholar] [CrossRef] [PubMed]
  154. Wolf, E.; Lin, C.Y.; Eilers, M.; Levens, D.L. Taming of the beast: Shaping myc-dependent amplification. Trends Cell Biol. 2015, 25, 241–248. [Google Scholar] [CrossRef] [Green Version]
  155. Korangath, P.; Teo, W.W.; Sadik, H.; Han, L.; Mori, N.; Huijts, C.M.; Wildes, F.; Bharti, S.; Zhang, Z.; Santa-Maria, C.A.; et al. Targeting glutamine metabolism in breast cancer with aminooxyacetate. Clin. Cancer Res. 2015, 21, 3263–3273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Shachaf, C.M.; Kopelman, A.M.; Arvanitis, C.; Karlsson, A.; Beer, S.; Mandl, S.; Bachmann, M.H.; Borowsky, A.D.; Ruebner, B.; Cardiff, R.D.; et al. Myc inactivation uncovers pluripotent differentiation and tumour dormancy in hepatocellular cancer. Nature 2004, 431, 1112–1117. [Google Scholar] [CrossRef] [PubMed]
  157. Ben-David, E.; Bester, A.C.; Shifman, S.; Kerem, B. Transcriptional dynamics in colorectal carcinogenesis: New insights into the role of c-myc and mir17 in benign to cancer transformation. Cancer Res. 2014, 74, 5532–5540. [Google Scholar] [CrossRef] [Green Version]
  158. Vita, M.; Henriksson, M. The Myc oncoprotein as a therapeutic target for human cancer. Semin. Cancer Biol. 2006, 16, 318–330. [Google Scholar] [CrossRef] [PubMed]
  159. Mossafa, H.; Damotte, D.; Jenabian, A.; Delarue, R.; Vincenneau, A.; Amouroux, I.; Jeandel, R.; Khoury, E.; Martelli, J.M.; Samson, T.; et al. Non-hodgkin’s lymphomas with burkitt-like cells are associated with c-myc amplification and poor prognosis. Leuk. Lymphoma 2006, 47, 1885–1893. [Google Scholar] [CrossRef] [PubMed]
  160. Pelengaris, S.; Khan, M.; Evan, G. C-myc: More than just a matter of life and death. Nat. Rev. Cancer 2002, 2, 764–776. [Google Scholar] [CrossRef] [PubMed]
  161. Schaub, F.X.; Dhankani, V.; Berger, A.C.; Trivedi, M.; Richardson, A.B.; Shaw, R.; Zhao, W.; Zhang, X.; Ventura, A.; Liu, Y.; et al. Pan-cancer alterations of the myc oncogene and its proximal network across the cancer genome atlas. Cell Syst. 2018, 6, 282–300.e282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Marinkovic, T.; Marinkovic, D. Obscure involvement of myc in neurodegenerative diseases and neuronal repair. Mol. Neurobiol. 2021, 58, 4169–4177. [Google Scholar] [CrossRef] [PubMed]
  163. Ferrer, I.; Blanco, R. N-myc and c-myc expression in alzheimer disease, huntington disease and parkinson disease. Brain Res. Mol. Brain Res. 2000, 77, 270–276. [Google Scholar] [CrossRef]
  164. Ferrer, I.; Blanco, R.; Carmona, M.; Puig, B. Phosphorylated c-myc expression in alzheimer disease, pick’s disease, progressive supranuclear palsy and corticobasal degeneration. Neuropathol. Appl. Neurobiol. 2001, 27, 343–351. [Google Scholar] [CrossRef]
  165. Blom, E.S.; Wang, Y.; Skoglund, L.; Hansson, A.C.; Ubaldi, M.; Lourdusamy, A.; Sommer, W.H.; Mielke, M.; Hyman, B.T.; Heilig, M.; et al. Increased mrna levels of tcf7l2 and myc of the wnt pathway in tg-arcswe mice and alzheimer’s disease brain. Int. J. Alzheimer Dis. 2010, 2011, 936580. [Google Scholar]
  166. Lee, H.G.; Casadesus, G.; Nunomura, A.; Zhu, X.; Castellani, R.J.; Richardson, S.L.; Perry, G.; Felsher, D.W.; Petersen, R.B.; Smith, M.A. The neuronal expression of myc causes a neurodegenerative phenotype in a novel transgenic mouse. Am. J. Pathol. 2009, 174, 891–897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Tao, L.; Zhu, Y.; Wang, R.; Han, J.; Ma, Y.; Guo, H.; Tang, W.; Zhuo, L.; Fan, Z.; Yin, A.; et al. N-myc downstream-regulated gene 2 deficiency aggravates memory impairment in alzheimer’s disease. Behav. Brain Res. 2020, 379, 112384. [Google Scholar] [CrossRef]
  168. Braicu, C.; Buse, M.; Busuioc, C.; Drula, R.; Gulei, D.; Raduly, L.; Rusu, A.; Irimie, A.; Atanasov, A.G.; Slaby, O.; et al. A comprehensive review on mapk: A promising therapeutic target in cancer. Cancers 2019, 11, 1618. [Google Scholar] [CrossRef] [Green Version]
  169. Cargnello, M.; Roux, P.P. Activation and function of the mapks and their substrates, the mapk-activated protein kinases. Microbiol. Mol. Biol. Rev. 2011, 75, 50–83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Lavoie, H.; Therrien, M. Regulation of raf protein kinases in erk signalling. Nat. Rev. Mol. Cell Biol. 2015, 16, 281–298. [Google Scholar] [CrossRef]
  171. Cerami, E.; Gao, J.; Dogrusoz, U.; Gross, B.E.; Sumer, S.O.; Aksoy, B.A.; Jacobsen, A.; Byrne, C.J.; Heuer, M.L.; Larsson, E.; et al. The cbio cancer genomics portal: An open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012, 2, 401–404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Gao, J.; Aksoy, B.A.; Dogrusoz, U.; Dresdner, G.; Gross, B.; Sumer, S.O.; Sun, Y.; Jacobsen, A.; Sinha, R.; Larsson, E.; et al. Integrative analysis of complex cancer genomics and clinical profiles using the cbioportal. Sci. Signal 2013, 6, pl1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Kim, E.K.; Choi, E.J. Pathological roles of mapk signaling pathways in human diseases. Biochim. Biophys. Acta 2010, 1802, 396–405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Pei, J.J.; Braak, H.; An, W.L.; Winblad, B.; Cowburn, R.F.; Iqbal, K.; Grundke-Iqbal, I. Up-regulation of mitogen-activated protein kinases erk1/2 and mek1/2 is associated with the progression of neurofibrillary degeneration in alzheimer’s disease. Brain Res. Mol. Brain Res. 2002, 109, 45–55. [Google Scholar] [CrossRef]
  175. Russo, C.; Dolcini, V.; Salis, S.; Venezia, V.; Zambrano, N.; Russo, T.; Schettini, G. Signal transduction through tyrosine-phosphorylated c-terminal fragments of amyloid precursor protein via an enhanced interaction with shc/grb2 adaptor proteins in reactive astrocytes of alzheimer’s disease brain. J. Biol. Chem. 2002, 277, 35282–35288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Kirouac, L.; Rajic, A.J.; Cribbs, D.H.; Padmanabhan, J. Activation of ras-erk signaling and gsk-3 by amyloid precursor protein and amyloid beta facilitates neurodegeneration in alzheimer’s disease. eNeuro 2017, 4. [Google Scholar] [CrossRef] [Green Version]
  177. Zhu, J.H.; Kulich, S.M.; Oury, T.D.; Chu, C.T. Cytoplasmic aggregates of phosphorylated extracellular signal-regulated protein kinases in lewy body diseases. Am. J. Pathol. 2002, 161, 2087–2098. [Google Scholar] [CrossRef] [Green Version]
  178. Sun, A.; Liu, M.; Nguyen, X.V.; Bing, G. P38 map kinase is activated at early stages in alzheimer’s disease brain. Exp. Neurol. 2003, 183, 394–405. [Google Scholar] [CrossRef]
  179. Hensley, K.; Floyd, R.A.; Zheng, N.Y.; Nael, R.; Robinson, K.A.; Nguyen, X.; Pye, Q.N.; Stewart, C.A.; Geddes, J.; Markesbery, W.R.; et al. P38 kinase is activated in the alzheimer’s disease brain. J. Neurochem. 1999, 72, 2053–2058. [Google Scholar] [CrossRef]
  180. Schnoder, L.; Hao, W.; Qin, Y.; Liu, S.; Tomic, I.; Liu, X.; Fassbender, K.; Liu, Y. Deficiency of neuronal p38alpha mapk attenuates amyloid pathology in alzheimer disease mouse and cell models through facilitating lysosomal degradation of bace1. J. Biol. Chem. 2016, 291, 2067–2079. [Google Scholar] [CrossRef] [Green Version]
  181. Pan, J.; Xiao, Q.; Sheng, C.Y.; Hong, Z.; Yang, H.Q.; Wang, G.; Ding, J.Q.; Chen, S.D. Blockade of the translocation and activation of c-jun n-terminal kinase 3 (jnk3) attenuates dopaminergic neuronal damage in mouse model of parkinson’s disease. Neurochem. Int. 2009, 54, 418–425. [Google Scholar] [CrossRef] [PubMed]
  182. Munoz, L.; Ralay Ranaivo, H.; Roy, S.M.; Hu, W.; Craft, J.M.; McNamara, L.K.; Chico, L.W.; Van Eldik, L.J.; Watterson, D.M. A novel p38 alpha mapk inhibitor suppresses brain proinflammatory cytokine up-regulation and attenuates synaptic dysfunction and behavioral deficits in an alzheimer’s disease mouse model. J. Neuroinflamm. 2007, 4, 21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Sclip, A.; Tozzi, A.; Abaza, A.; Cardinetti, D.; Colombo, I.; Calabresi, P.; Salmona, M.; Welker, E.; Borsello, T. C-jun n-terminal kinase has a key role in alzheimer disease synaptic dysfunction in vivo. Cell Death Dis. 2014, 5, e1019. [Google Scholar] [CrossRef] [PubMed]
  184. Du, Y.; Du, Y.; Zhang, Y.; Huang, Z.; Fu, M.; Li, J.; Pang, Y.; Lei, P.; Wang, Y.T.; Song, W.; et al. Mkp-1 reduces abeta generation and alleviates cognitive impairments in alzheimer’s disease models. Signal Transduct. Target Ther. 2019, 4, 58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Zhou, Q.; Wang, M.; Du, Y.; Zhang, W.; Bai, M.; Zhang, Z.; Li, Z.; Miao, J. Inhibition of c-jun n-terminal kinase activation reverses alzheimer disease phenotypes in appswe/ps1de9 mice. Ann. Neurol. 2015, 77, 637–654. [Google Scholar] [CrossRef] [PubMed]
  186. Bohush, A.; Niewiadomska, G.; Filipek, A. Role of mitogen activated protein kinase signaling in parkinson’s disease. Int. J. Mol. Sci. 2018, 19, 2973. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Rai, S.N.; Dilnashin, H.; Birla, H.; Singh, S.S.; Zahra, W.; Rathore, A.S.; Singh, B.K.; Singh, S.P. The role of pi3k/akt and erk in neurodegenerative disorders. Neurotox. Res. 2019, 35, 775–795. [Google Scholar] [CrossRef] [PubMed]
  188. Correa, S.A.; Eales, K.L. The role of p38 mapk and its substrates in neuronal plasticity and neurodegenerative disease. J. Signal Transduct. 2012, 2012, 649079. [Google Scholar] [CrossRef] [Green Version]
  189. Fan, J.; Gladding, C.M.; Wang, L.; Zhang, L.Y.; Kaufman, A.M.; Milnerwood, A.J.; Raymond, L.A. P38 mapk is involved in enhanced nmda receptor-dependent excitotoxicity in yac transgenic mouse model of huntington disease. Neurobiol. Dis. 2012, 45, 999–1009. [Google Scholar] [CrossRef]
  190. Apostol, B.L.; Illes, K.; Pallos, J.; Bodai, L.; Wu, J.; Strand, A.; Schweitzer, E.S.; Olson, J.M.; Kazantsev, A.; Marsh, J.L.; et al. Mutant huntingtin alters mapk signaling pathways in pc12 and striatal cells: Erk1/2 protects against mutant huntingtin-associated toxicity. Hum. Mol. Genet. 2006, 15, 273–285. [Google Scholar] [CrossRef] [Green Version]
  191. Lane, D.P. Cancer. P53, guardian of the genome. Nature 1992, 358, 15–16. [Google Scholar] [CrossRef] [PubMed]
  192. Levine, A.J. P53: 800 million years of evolution and 40 years of discovery. Nat. Rev. Cancer 2020, 20, 471–480. [Google Scholar] [CrossRef] [PubMed]
  193. Marei, H.E.; Althani, A.; Afifi, N.; Hasan, A.; Caceci, T.; Pozzoli, G.; Morrione, A.; Giordano, A.; Cenciarelli, C. P53 signaling in cancer progression and therapy. Cancer Cell Int. 2021, 21, 703. [Google Scholar] [CrossRef] [PubMed]
  194. Muller, P.A.; Vousden, K.H. Mutant p53 in cancer: New functions and therapeutic opportunities. Cancer Cell 2014, 25, 304–317. [Google Scholar] [CrossRef] [Green Version]
  195. Xu, J.; Reumers, J.; Couceiro, J.R.; De Smet, F.; Gallardo, R.; Rudyak, S.; Cornelis, A.; Rozenski, J.; Zwolinska, A.; Marine, J.C.; et al. Gain of function of mutant p53 by coaggregation with multiple tumor suppressors. Nat. Chem. Biol. 2011, 7, 285–295. [Google Scholar] [CrossRef] [Green Version]
  196. Kim, M.P.; Lozano, G. Mutant p53 partners in crime. Cell Death Differ. 2018, 25, 161–168. [Google Scholar] [CrossRef]
  197. D’Orazi, G.; Cirone, M. Mutant p53 and cellular stress pathways: A criminal alliance that promotes cancer progression. Cancers 2019, 11, 614. [Google Scholar] [CrossRef] [Green Version]
  198. Szybinska, A.; Lesniak, W. P53 dysfunction in neurodegenerative diseases—The cause or effect of pathological changes? Aging Dis. 2017, 8, 506–518. [Google Scholar] [CrossRef] [Green Version]
  199. Chang, J.R.; Ghafouri, M.; Mukerjee, R.; Bagashev, A.; Chabrashvili, T.; Sawaya, B.E. Role of p53 in neurodegenerative diseases. Neurodegener. Dis. 2012, 9, 68–80. [Google Scholar] [CrossRef] [Green Version]
  200. Kitamura, Y.; Shimohama, S.; Kamoshima, W.; Matsuoka, Y.; Nomura, Y.; Taniguchi, T. Changes of p53 in the brains of patients with alzheimer’s disease. Biochem. Biophys. Res. Commun. 1997, 232, 418–421. [Google Scholar] [CrossRef]
  201. Chung, Y.H.; Shin, C.; Kim, M.J.; Lee, B.; Park, K.H.; Cha, C.I. Immunocytochemical study on the distribution of p53 in the hippocampus and cerebellum of the aged rat. Brain Res. 2000, 885, 137–141. [Google Scholar] [CrossRef]
  202. Tatton, N.A. Increased caspase 3 and bax immunoreactivity accompany nuclear gapdh translocation and neuronal apoptosis in parkinson’s disease. Exp. Neurol. 2000, 166, 29–43. [Google Scholar] [CrossRef] [PubMed]
  203. Sajan, F.D.; Martiniuk, F.; Marcus, D.L.; Frey, W.H., 2nd; Hite, R.; Bordayo, E.Z.; Freedman, M.L. Apoptotic gene expression in alzheimer’s disease hippocampal tissue. Am. J. Alzheimer Dis. Other Dement. 2007, 22, 319–328. [Google Scholar] [CrossRef] [PubMed]
  204. Farmer, K.M.; Ghag, G.; Puangmalai, N.; Montalbano, M.; Bhatt, N.; Kayed, R. P53 aggregation, interactions with tau, and impaired DNA damage response in alzheimer’s disease. Acta Neuropathol. Commun. 2020, 8, 132. [Google Scholar] [CrossRef] [PubMed]
  205. Alves da Costa, C.; Sunyach, C.; Pardossi-Piquard, R.; Sevalle, J.; Vincent, B.; Boyer, N.; Kawarai, T.; Girardot, N.; St George-Hyslop, P.; Checler, F. Presenilin-dependent gamma-secretase-mediated control of p53-associated cell death in alzheimer’s disease. J. Neurosci. 2006, 26, 6377–6385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Pastorcic, M.; Das, H.K. Regulation of transcription of the human presenilin-1 gene by ets transcription factors and the p53 protooncogene. J. Biol. Chem. 2000, 275, 34938–34945. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Bae, B.I.; Xu, H.; Igarashi, S.; Fujimuro, M.; Agrawal, N.; Taya, Y.; Hayward, S.D.; Moran, T.H.; Montell, C.; Ross, C.A.; et al. P53 mediates cellular dysfunction and behavioral abnormalities in huntington’s disease. Neuron 2005, 47, 29–41. [Google Scholar] [CrossRef] [Green Version]
  208. Ryan, A.B.; Zeitlin, S.O.; Scrable, H. Genetic interaction between expanded murine hdh alleles and p53 reveal deleterious effects of p53 on huntington’s disease pathogenesis. Neurobiol. Dis. 2006, 24, 419–427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  209. Qi, X.; Davis, B.; Chiang, Y.H.; Filichia, E.; Barnett, A.; Greig, N.H.; Hoffer, B.; Luo, Y. Dopaminergic neuron-specific deletion of p53 gene is neuroprotective in an experimental parkinson’s disease model. J. Neurochem. 2016, 138, 746–757. [Google Scholar] [CrossRef] [Green Version]
  210. Otto, T.; Sicinski, P. Cell cycle proteins as promising targets in cancer therapy. Nat. Rev. Cancer 2017, 17, 93–115. [Google Scholar] [CrossRef] [Green Version]
  211. Xiao, Y.; Dong, J. The hippo signaling pathway in cancer: A cell cycle perspective. Cancers 2021, 13, 6214. [Google Scholar] [CrossRef] [PubMed]
  212. van Leeuwen, L.A.; Hoozemans, J.J. Physiological and pathophysiological functions of cell cycle proteins in post-mitotic neurons: Implications for alzheimer’s disease. Acta Neuropathol. 2015, 129, 511–525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Ippati, S.; Deng, Y.; van der Hoven, J.; Heu, C.; van Hummel, A.; Chua, S.W.; Paric, E.; Chan, G.; Feiten, A.; Fath, T.; et al. Rapid initiation of cell cycle reentry processes protects neurons from amyloid-beta toxicity. Proc. Natl. Acad. Sci. USA 2021, 118, e2011876118. [Google Scholar] [CrossRef]
  214. Joseph, C.; Mangani, A.S.; Gupta, V.; Chitranshi, N.; Shen, T.; Dheer, Y.; Kb, D.; Mirzaei, M.; You, Y.; Graham, S.L.; et al. Cell cycle deficits in neurodegenerative disorders: Uncovering molecular mechanisms to drive innovative therapeutic development. Aging Dis. 2020, 11, 946–966. [Google Scholar] [CrossRef] [PubMed]
  215. Yang, Y.; Mufson, E.J.; Herrup, K. Neuronal cell death is preceded by cell cycle events at all stages of alzheimer’s disease. J. Neurosci. 2003, 23, 2557–2563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Crews, L.; Patrick, C.; Adame, A.; Rockenstein, E.; Masliah, E. Modulation of aberrant cdk5 signaling rescues impaired neurogenesis in models of alzheimer’s disease. Cell Death Dis. 2011, 2, e120. [Google Scholar] [CrossRef] [PubMed]
  217. Pelegri, C.; Duran-Vilaregut, J.; del Valle, J.; Crespo-Biel, N.; Ferrer, I.; Pallas, M.; Camins, A.; Vilaplana, J. Cell cycle activation in striatal neurons from huntington’s disease patients and rats treated with 3-nitropropionic acid. Int. J. Dev. Neurosci. 2008, 26, 665–671. [Google Scholar] [CrossRef] [PubMed]
  218. Manickam, N.; Radhakrishnan, R.K.; Vergil Andrews, J.F.; Selvaraj, D.B.; Kandasamy, M. Cell cycle re-entry of neurons and reactive neuroblastosis in huntington’s disease: Possibilities for neural-glial transition in the brain. Life Sci. 2020, 263, 118569. [Google Scholar] [CrossRef]
  219. Park, K.H.; Hallows, J.L.; Chakrabarty, P.; Davies, P.; Vincent, I. Conditional neuronal simian virus 40 t antigen expression induces alzheimer-like tau and amyloid pathology in mice. J. Neurosci. 2007, 27, 2969–2978. [Google Scholar] [CrossRef]
  220. Smith, P.D.; Crocker, S.J.; Jackson-Lewis, V.; Jordan-Sciutto, K.L.; Hayley, S.; Mount, M.P.; O’Hare, M.J.; Callaghan, S.; Slack, R.S.; Przedborski, S.; et al. Cyclin-dependent kinase 5 is a mediator of dopaminergic neuron loss in a mouse model of parkinson’s disease. Proc. Natl. Acad. Sci. USA 2003, 100, 13650–13655. [Google Scholar] [CrossRef] [Green Version]
  221. Stefanis, L. Alpha-synuclein in parkinson’s disease. Cold Spring Harb. Perspect. Med. 2012, 2, a009399. [Google Scholar] [CrossRef] [Green Version]
  222. Lee, S.S.; Kim, Y.M.; Junn, E.; Lee, G.; Park, K.H.; Tanaka, M.; Ronchetti, R.D.; Quezado, M.M.; Mouradian, M.M. Cell cycle aberrations by alpha-synuclein over-expression and cyclin b immunoreactivity in lewy bodies. Neurobiol. Aging 2003, 24, 687–696. [Google Scholar] [CrossRef]
  223. Janku, F. Phosphoinositide 3-kinase (pi3k) pathway inhibitors in solid tumors: From laboratory to patients. Cancer Treat. Rev. 2017, 59, 93–101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Thorpe, L.M.; Yuzugullu, H.; Zhao, J.J. Pi3k in cancer: Divergent roles of isoforms, modes of activation and therapeutic targeting. Nat. Rev. Cancer 2015, 15, 7–24. [Google Scholar] [CrossRef]
  225. Janku, F.; Yap, T.A.; Meric-Bernstam, F. Targeting the pi3k pathway in cancer: Are we making headway? Nat. Rev. Clin. Oncol. 2018, 15, 273–291. [Google Scholar] [CrossRef] [PubMed]
  226. Zhang, Y.; Kwok-Shing Ng, P.; Kucherlapati, M.; Chen, F.; Liu, Y.; Tsang, Y.H.; de Velasco, G.; Jeong, K.J.; Akbani, R.; Hadjipanayis, A.; et al. A pan-cancer proteogenomic atlas of pi3k/akt/mtor pathway alterations. Cancer Cell 2017, 31, 820–832.e823. [Google Scholar] [CrossRef] [Green Version]
  227. Keniry, M.; Parsons, R. The role of pten signaling perturbations in cancer and in targeted therapy. Oncogene 2008, 27, 5477–5485. [Google Scholar] [CrossRef] [Green Version]
  228. Long, H.Z.; Cheng, Y.; Zhou, Z.W.; Luo, H.Y.; Wen, D.D.; Gao, L.C. Pi3k/akt signal pathway: A target of natural products in the prevention and treatment of alzheimer’s disease and parkinson’s disease. Front. Pharmacol. 2021, 12, 648636. [Google Scholar] [CrossRef]
  229. Heras-Sandoval, D.; Perez-Rojas, J.M.; Hernandez-Damian, J.; Pedraza-Chaverri, J. The role of pi3k/akt/mtor pathway in the modulation of autophagy and the clearance of protein aggregates in neurodegeneration. Cell Signal 2014, 26, 2694–2701. [Google Scholar] [CrossRef] [PubMed]
  230. Curtis, D.; Bandyopadhyay, S. Mini-review: Role of the pi3k/akt pathway and tyrosine phosphatases in alzheimer’s disease susceptibility. Ann. Hum. Genet. 2021, 85, 1–6. [Google Scholar] [CrossRef]
  231. Griffin, R.J.; Moloney, A.; Kelliher, M.; Johnston, J.A.; Ravid, R.; Dockery, P.; O’Connor, R.; O’Neill, C. Activation of akt/pkb, increased phosphorylation of akt substrates and loss and altered distribution of akt and pten are features of alzheimer’s disease pathology. J. Neurochem. 2005, 93, 105–117. [Google Scholar] [CrossRef] [PubMed]
  232. Timmons, S.; Coakley, M.F.; Moloney, A.M.; O’Neill, C. Akt signal transduction dysfunction in parkinson’s disease. Neurosci. Lett. 2009, 467, 30–35. [Google Scholar] [CrossRef]
  233. Greene, L.A.; Levy, O.; Malagelada, C. Akt as a victim, villain and potential hero in parkinson’s disease pathophysiology and treatment. Cell Mol. Neurobiol. 2011, 31, 969–978. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  234. Malagelada, C.; Jin, Z.H.; Greene, L.A. Rtp801 is induced in parkinson’s disease and mediates neuron death by inhibiting akt phosphorylation/activation. J. Neurosci. 2008, 28, 14363–14371. [Google Scholar] [CrossRef] [Green Version]
  235. Luo, S.; Kang, S.S.; Wang, Z.H.; Liu, X.; Day, J.X.; Wu, Z.; Peng, J.; Xiang, D.; Springer, W.; Ye, K. Akt phosphorylates nqo1 and triggers its degradation, abolishing its antioxidative activities in parkinson’s disease. J. Neurosci. 2019, 39, 7291–7305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  236. Ries, V.; Henchcliffe, C.; Kareva, T.; Rzhetskaya, M.; Bland, R.; During, M.J.; Kholodilov, N.; Burke, R.E. Oncoprotein akt/pkb induces trophic effects in murine models of parkinson’s disease. Proc. Natl. Acad. Sci. USA 2006, 103, 18757–18762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  237. Aleyasin, H.; Rousseaux, M.W.; Marcogliese, P.C.; Hewitt, S.J.; Irrcher, I.; Joselin, A.P.; Parsanejad, M.; Kim, R.H.; Rizzu, P.; Callaghan, S.M.; et al. Dj-1 protects the nigrostriatal axis from the neurotoxin mptp by modulation of the akt pathway. Proc. Natl. Acad. Sci. USA 2010, 107, 3186–3191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  238. Hu, M.; Li, F.; Wang, W. Vitexin protects dopaminergic neurons in mptp-induced parkinson’s disease through pi3k/akt signaling pathway. Drug Des. Dev. Ther. 2018, 12, 565–573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  239. Tramutola, A.; Triplett, J.C.; Di Domenico, F.; Niedowicz, D.M.; Murphy, M.P.; Coccia, R.; Perluigi, M.; Butterfield, D.A. Alteration of mtor signaling occurs early in the progression of alzheimer disease (ad): Analysis of brain from subjects with pre-clinical ad, amnestic mild cognitive impairment and late-stage ad. J. Neurochem. 2015, 133, 739–749. [Google Scholar] [CrossRef] [PubMed]
  240. Kitagishi, Y.; Nakanishi, A.; Ogura, Y.; Matsuda, S. Dietary regulation of pi3k/akt/gsk-3beta pathway in alzheimer’s disease. Alzheimer Res. Ther. 2014, 6, 35. [Google Scholar] [CrossRef] [Green Version]
  241. Mallorqui-Bague, N.; Lozano-Madrid, M.; Toledo, E.; Corella, D.; Salas-Salvado, J.; Cuenca-Royo, A.; Vioque, J.; Romaguera, D.; Martinez, J.A.; Warnberg, J.; et al. Type 2 diabetes and cognitive impairment in an older population with overweight or obesity and metabolic syndrome: Baseline cross-sectional analysis of the predimed-plus study. Sci. Rep. 2018, 8, 16128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  242. De Groot, N.S.; Burgas, M.T. Is membrane homeostasis the missing link between inflammation and neurodegenerative diseases? Cell Mol. Life Sci. 2015, 72, 4795–4805. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  243. Hu, J.; Wang, X. Alzheimer’s disease: From pathogenesis to mesenchymal stem cell therapy—Bridging the missing link. Front. Cell. Neurosci. 2022, 15, 811852. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Cellular functions altered in cancer and neurodegenerative diseases. Alterations of the same molecular mechanisms can drive cell survival and proliferation in cancer and cell death and apoptosis in the development of different neurodegenerative disease.
Figure 1. Cellular functions altered in cancer and neurodegenerative diseases. Alterations of the same molecular mechanisms can drive cell survival and proliferation in cancer and cell death and apoptosis in the development of different neurodegenerative disease.
Ijms 23 03223 g001
Table 1. Brain areas and neuronal populations affected by neurodegenerative disorders.
Table 1. Brain areas and neuronal populations affected by neurodegenerative disorders.
Neurodegenerative
Disease
CNS AreaNeuronal PopulationREF
Alzheimer’s diseaseHippocampus (CA1).
Entorhinal Cortex.
Locus coeruleus.
Basal forebrain.
Pyramidal neurons.
Cholinergic neurons.
Noradrenergic neurons.
[14,15,16,17,18,19]
Parkinson’s diseaseSubstantia nigra pars compacta (SNpc).
VTA (lower levels of
Degeneration)
Dopaminergic neurons[20,21,22,23]
Huntington’s DiseaseStriatumMedium spiny GABAergic neurons (MSN).[24,25]
Amyotrophic lateral
Sclerosis (ALS)
Spinal cord.
Motor cortex.
Brain stem.
Motor neurons[26,27,28]
Pick’s DiseaseHippocampus
Amygdala.
Frontal and temporal lobes
Pyramidal and granular neurons.[29,30,31,32]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Varela, L.; Garcia-Rendueles, M.E.R. Oncogenic Pathways in Neurodegenerative Diseases. Int. J. Mol. Sci. 2022, 23, 3223. https://doi.org/10.3390/ijms23063223

AMA Style

Varela L, Garcia-Rendueles MER. Oncogenic Pathways in Neurodegenerative Diseases. International Journal of Molecular Sciences. 2022; 23(6):3223. https://doi.org/10.3390/ijms23063223

Chicago/Turabian Style

Varela, Luis, and Maria E. R. Garcia-Rendueles. 2022. "Oncogenic Pathways in Neurodegenerative Diseases" International Journal of Molecular Sciences 23, no. 6: 3223. https://doi.org/10.3390/ijms23063223

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