Home Life Sciences Unravelling the genetic links between Parkinson’s disease and lung cancer
Article Open Access

Unravelling the genetic links between Parkinson’s disease and lung cancer

  • Yong Qi Leong

    Yong Qi Leong is currently in her second year of PhD in Monash University Malaysia. She is awarded with Graduate Research Merit Scholarship offered by Monash University Malaysia. Her research focuses on molecular biology, lung cancer studies and protein–protein interactions. She has four publications throughout her research journey. Also, she is the executive committee of BioMedKL and holds several projects in order to contribute in the community.

    , Rhun Yian Koh

    Dr. Rhun Yian Koh obtained her BSc Biomedical Science (Hons) and PhD (Immunobiology) degrees at the University Putra Malaysia. She is currently a Senior Lecturer of the Division of Applied Biomedical Science, International Medical University. Her research interests include natural product and neuroscience. She has received multiple national and internal research grants for the past few years, and published more than 80 articles. She has also presented her research findings in multiple conferences. She is a member of the Malaysian Biomedical Science Association.

    , Soi Moi Chye

    Associate Professor Dr. Soi Moi Chye graduated from Kaohsiung Medical University, Taiwan, PhD. She has more than 20 years of teaching experience in universities in Taiwan and Malaysia. Her research interests include neuroscience, toxicology, cancer and immunodiagnostic of parasite infection. At present, she has published more than 77 peer-reviewed articles, her RG score is 32.78, and her h-index is 19.

    and Khuen Yen Ng

    Dr. Khuen Yen Ng completed his BSc in Monash University. He then pursued his MSc study in University of New South Wales. Later he undertook his PhD study in Neuroscience in the prestigious Neuroscience Research Australia. His research interest includes: neurodegenerative diseases (including Parksinons’s disease and Alzheimer’s disease), oncology and sleep studies. He is the current advisor of Malaysian Parkinson’s Disease Association and he has published around 50 articles, with h-index of 12.

    ORCID logo EMAIL logo
Published/Copyright: January 13, 2023

Abstract

Increase evidence from epidemiological studies have shown an inverse association between Parkinson’s disease (PD) and lung cancer. PD and lung cancer are both geriatric diseases, where these two diseases are sharing some common genetic determinants. Several PD-associated genes including alpha synuclein (SNCA), PTEN-induced kinase 1 (PINK1), parkin, parkinsonism associated deglycase (DJ-1), leucine-rich repeat kinase 2 (LRRK2), F-box protein 7 (FBXO7) and ubiquitin C-terminal hydrolase L1 (UCHL1) were reported to have altered expressions in lung cancer patients. This indicates that certain PD-associated genes might be important in conferring anticancer effects. This review aims to depict the physiological functions of these genes, and discuss the putative roles of these PD-associated genes in lung cancer. The understanding of the roles of these genes in the lung cancer progression might be important in the identification of new treatment targets for lung cancer. Gene therapy that aims to alter the expressions of these genes could be developed for future anticancer therapy. As a result, studying the roles of these genes in lung cancer may also help to understand their involvements as well as their roles in the pathogenesis of PD.

Introduction

In the past 30 years, accumulating evidence have reported a negative association between Parkinson’s disease (PD) and lung cancer (Becker et al. 2010; Freedman et al. 2016; Park et al. 2019a). In the latest meta-analysis conducted by our group, the diagnosis of PD was associated with a 44% reduced risk of lung cancer (Leong et al. 2021). PD is the second most common neurodegenerative disease that is affecting 1% of the population above 60 years (Tysnes and Storstein 2017). The pathological hallmark of this disease is the loss of dopaminergic neurons in the substantia nigra (Jankovic 2008). PD is characterized by deteriorated motor features including tremors, muscular rigidity, postural imbalance and bradykinesia and a range of non-motor symptoms such as autonomic, cognitive and behavioral dysfunctions (Kalia and Lang 2015; Postuma et al. 2015). Etiology of PD is unknown but age is known to be the major risk factor for PD. Evidence shows that multiple cellular events such as misfolded proteins aggregations, oxidative stress, neuroinflammation, genetic mutations and protein clearance interruption are in place to trigger the neurodegeneration in PD (Maiti et al. 2017). Studies showed that 5–10% of both autosomal and inherited forms of PD are linked to genetic factors (Klein and Westenberger 2012; Maiti et al. 2017). The most common causative genes of PD include SNCA, parkin, DJ-1, PINK1, UCHL1 and LRRK2 (Deng et al. 2018; Klein and Westenberger 2012; Maiti et al. 2017; Selvaraj and Piramanayagam 2019).

Other than that, lung cancer is a highly invasive and rapidly metastasizing cancer. It is known to be one of the leading causes of death attributed to cancer worldwide. Lung cancer accounted for 18.4% of the total cancer deaths in 2018, with an estimation of 1.8 million deaths worldwide (Bray et al. 2018). It is the leading cause of mortality in patients aged between 40 and 80 years old (Howlader et al. 2014). Although lung cancer treatments have been improved in the past decades, lung cancer prognosis is still poor, and the 5 year survival rate remains low (Allemani et al. 2018). Lung cancer is classified into two broad categories, which are non-small cell lung carcinoma (NSCLC) and small cell lung cancer (SCLC) (Testa et al. 2018). 15 percent of lung cancers are represented by SCLC, an aggressively malignant tumor exhibiting neuroendocrine characteristics. SCLC is found to be strongly associated with smoking (Furrukh 2013). On the other hand, 85% of lung cancer cases are represented by NSCLC and it can be further categorized into adenocarcinoma (40%), squamous cell carcinoma (20–30%) and large cell carcinoma (5–10%) (Zappa and Mousa 2016). Most of the NSCLC patients are diagnosed at an advanced stage with median overall survival ranges between 7 and 12 months, depending on the histology types and therapeutic strategies (Schad et al. 2018). Lung adenocarcinoma usually arises in the glandular epithelium of lung parenchyma consisting of type II pneumocytes whereas lung squamous cell carcinoma originates from the basal cells in the central airways (Pikor et al. 2013; Travis et al. 2011). In contrast, large cell carcinoma is an undifferentiated tumor harboring neuroendocrine features (Pikor et al. 2013).

Previously, lung cancer and PD are two diseases thought to be unrelated to each other. However, recent emerging evidence suggests that they not only share some common risk factors such as aging, but also share some common genetic features. For instance, some PD-associated genes were also reported in the pathology of lung cancer (Lebovitz et al. 2021; Liu et al. 2015). A study by Liu et al. identified elevated expressions of these PD-associated genes in NSCLC patients where the tumor samples were collected at the time of surgical resection (Liu et al. 2015). Therefore, this review aims to depict the physiological functions of PD-associated genes, followed by in-depth discussions on the putative roles of these genes in lung cancer with a focus on the molecular connections between lung cancer and PD (Table 1).

Table 1:

PD-associated genes and their putative roles identified in lung cancer.

Gene Gene locus Expression in lung cancer Putative roles in lung cancer
SNCA Chromosome 4q21.2–q22 (Kim et al. 2014; Oczkowska et al. 2013) Downregulation EGFR trafficking
Bcl-2 regulated apoptosisa
Parkin Chromosome 6q25–q27 (Cesari et al. 2003; Quinn et al. 2020) Upregulation Cell cycle progression (cyclin E ubiquitination) (Park et al. 2019b; Veeriah et al. 2010)
EGFR trafficking and PI3K-Akt pathway regulation (Eps 15 ubiquitination) (Fallon et al. 2006; Husnjak and Dikic 2006)
Mitophagy (PHB2 regulation)
PINK1 Chromosome 1p36.12 (Quinn et al. 2020) Upregulation Cell migration and proliferation (mitophagy) (Lu et al. 2020)
Cell survival and proliferation (NF-κB pathway regulation) (Zhang et al. 2017)
Bcl-2 regulated apoptosis (Liu et al. 2018)
DJ-1 Chromosome 1p36.23 (Zhang et al. 2020b) Upregulation PI3k-Akt pathway regulation (antagonist of PTEN) (Kim et al. 2005a; Yang et al. 2005)
p53-mediated apoptosis (Shinbo et al. 2005; Vasseur et al. 2009)
FBXO7 Chromosome 22q12–q13 (Conedera et al. 2016) Upregulation Cell cycle progression and cell proliferation (cyclin D-CDK6 formation) (Laman 2006; Laman et al. 2005)
Cell cycle progression (NF-κB pathway regulation via cIAP1) (Chang et al. 2006; Kuiken et al. 2012)
UCHL1 Chromosome 4p14 (Ragland et al. 2009) Upregulation Cell proliferation (deubiquitination of cyclins)a (MAPK and PI3K-Akt pathways activation) (Hurst-Kennedy et al. 2012; Kim et al. 2009)
LRRK2 Chromosome 12p11.2–q13.1 (Zimprich et al. 2004) Downregulation ERK and JNK dependent autophagy (Herzig et al. 2011; Tian et al. 2021)
  1. Elevated expressions of parkin, PINK1, DJ-1, FBXO7 and UCHL1 and downregulation of SNCA and LRRK2 were detected in lung cancer. These genes regulate cell proliferation, cell survival, apoptosis, mitophagy/autophagy and cell cycle progression via various signaling pathways in order to promote lung carcinogenesis. Abbreviations: SNCA, alpha synuclein; PINK1, PTEN-induced putative kinase one; FBXO7, F-box protein 7; UCHL1, ubiquitin carboxyl-terminal esterase L1; LRRK2, leucine-rich repeat kinase two; EGFR, epidermal growth factor receptor; Bcl-2, B-cell lymphoma two; PI3k-Akt, phosphatidylinositol 3-kinase-protein kinase B; Eps 15, epidermal growth factor receptor pathway substrate 15; PHB2, prohibitin two; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; PTEN, phosphatase and tensin homolog; CDK6, cyclin-dependent kinase 6; CIAP1, cellular inhibitor of apoptosis one; ERK, extracellular signal-regulated kinase; JNK, c-Jun N-terminal kinases. aPutative roles remain speculative.

Current therapeutic strategies for lung cancer and their drawbacks

The current therapies for lung cancer include surgery, radiation therapy, chemotherapy, targeted therapy, immunotherapy or combined modality approach. Surgery is the standard of care for early-stage lung cancer patients (Dómine et al. 2020; Raman et al. 2018). Radiotherapy is another option for the early-stage patients, which damages DNA within cancerous cells (Zappa and Mousa 2016). For advanced-stage patients, a regimen of platinum-based chemotherapy (cisplatin or carboplatin) in combination with gemcitabine, pemetrexed, irinotecan or docetaxel is administered for a total of 4–6 cycles as first-line treatment for both NSCLC and SCLC patients (Chan and Coward 2013; Masters et al. 2015). However, the combinations of chemotherapy and radiotherapy only confer modest benefit in prolonging the overall survival as well as reducing the adverse events (Dómine et al. 2020; Zappa and Mousa 2016).

Targeted therapies involve the use of molecular inhibitors to block specific signaling pathways specifically. For instance, gefitinib, erlotinib and ceritinib which belong to the drug class of epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs), are the reversible competitors of ATP that bind to EGFR tyrosine kinase domain. These inhibitors are used to treat NSCLC patients with EGFR mutations with either L858R substitutions in exon 21 or amino acid deletions in exon 19 (Abidin et al. 2010; Rothschild 2014; Testa et al. 2018). Another FDA-approved agent known as crizotinib is an inhibitor of ALK, MET and ROS tyrosine kinases (Kazandjian et al. 2014; Solomon et al. 2014). These treatment strategies are promising as they confer progression-free survival longer than chemotherapy (Li et al. 2008; Richer et al. 2015; Rothschild 2014). Unfortunately, these therapies have some drawbacks, for instance, targeted therapies often result in gain-of-function mutations that lead to additional drug resistance characteristics in cancer cells. For example, the EGFR T790M mutation in exon 20, which resulted from prolonged EGFR TKI therapy, was known to disrupt the inhibitory activities of the TKIs (Kobayashi et al. 2005; Sun et al. 2013). Resistance to first-and second-generation EGFR TKIs which is caused by this mutation can be overcome by several third-generation irreversible EGFR TKIs such as osimertinib (Mok et al. 2017; Soria et al. 2018). Nevertheless, acquired resistance to osimertinib has been reported in several cases. Furthermore, a broad spectrum of drug resistance including KRAS, ALK, MET, cKIT amplification, HER1, HER2 and HER3 upregulation and L1196M mutations has been discovered in NSCLC patients (Chen et al. 2013; Doebele et al. 2012; Papadimitrakopoulou et al. 2018; Tanizaki et al. 2012). Similar drawback was observed in the SCLC patients treated with inhibitors such as gefitinib and imatinib, with no improvement in survival rate observed in small phase II trials (Abidin et al. 2010).

On top of that, immunotherapy boosts the body’s natural defense systems to defeat cancer by targeting immune checkpoint pathways (Onoi et al. 2020). Cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), programmed death 1 (PD-1) and its ligand (PD-L1) are the immune checkpoint inhibitors that block the immunosuppressive mechanisms in the lung cancer cells (Kim and Choi 2020; Saltos et al. 2020). Clinical trials have revealed that immunotherapy significantly improves lung cancer patients’ progression-free survival and overall survival (Kim and Choi 2020; Saltos et al. 2020). However, immune checkpoint inhibitors may cause immune-related adverse events which are resulted from non-specific activation of the immune system (Puzanov et al. 2017). This results in the high toxicities which could affect various organs including the lungs, liver, nervous system and thyroid in the patients who receiving the therapy (Kim and Choi 2020; Saltos et al. 2020).

Although there are many therapeutic agents used in the lung cancer treatment, their long-term outcomes still require further improvements. The 5 year survival rate of lung cancer patients remains low (15–20%), although it has been improved (Chen et al. 2020; Garon et al. 2019). Numerous therapeutic agents undergoing clinical trials seem to show promising results, however, the outcomes are immature and the optimal dosage and sequences are yet to be determined (Chen et al. 2020; Majeed et al. 2021). Furthermore, it is known that undesirable drug resistance and toxicity are inevitable (Chan and Hughes 2015). Disease progression has been another challenge for lung cancer therapies since lung cancer prognosis remains disappointing, with a 5 year survival rate of only approximately 15% (Edwards et al. 2014). Hence, a better understanding on the roles of these PD-associated genes and identifying the escape pathways in lung cancer could be significant in discovering new treatment strategies for lung cancer.

SNCA

SNCA encodes α-synuclein which is a 140 amino acid protein that consists of three distinct structural domains including an N-terminal region (residues 1–60), a central hydrophobic region (residues 61–95) and a C-terminal region (residues 96–140) (Kim et al. 2014; Snead and Eliezer 2014). α-Synuclein is abundantly found within the Lewy bodies, the pathological hallmark of PD, as amyloid fibrils (Kim et al. 2014; Spillantini 1999). Several variants with missense mutations (A53T, E46K and A30P, H50Q and G51D) have been identified in the cases of familial PD (Appel-Cresswell et al. 2013; Lesage et al. 2013; Proukakis et al. 2013). These mutations are believed to either accelerate the aggregation rate of fibrils formation or change the conformation or oligomerization upon aggregations (Meade et al. 2019; Snead and Eliezer 2014). Physiological functions of α-synuclein have been related to the regulation of synaptic vesicles for the release of neurotransmitters such as dopamine (Butler et al. 2017; Janezic et al. 2013) and the regulation of endoplasmic reticulum-Golgi transport (Cooper et al. 2006). Overexpression of SNCA has been reported in PD (Stefanis 2012; Tagliafierro and Chiba-Falek 2016). Aggregates of α-synuclein, especially the soluble oligomeric form, exert neurotoxicity that precedes synaptic dysfunction and ultimately cause neuronal cell death (Ingelsson 2016).

In PD, overexpression of wild-type or mutated SNCA triggered mitochondrial-mediated apoptosis that leads to neuronal cell death. This mechanism is thought to be mediated by the upregulation of Bax and downregulation of Bcl-xL as a result of interaction between SNCA and Bad, a pro-apoptotic member of the Bcl-family (Ahmad et al. 2007; Seo et al. 2002). Bad initiates allosteric activation of Bax to promote conformational change, resulting in cytochrome c release and caspases activation (Ahmad et al. 2007; Seo et al. 2002). Other than that, SNCA activates phosphatidylinositol 3-kinase-protein kinase B (PI3K-Akt) at a low expression level and subsequently increases expressions of anti-apoptotic Bcl-family members in neuronal cells. PI3K-Akt phosphorylates Akt, which activates Bad phosphorylation, thereby reducing interaction between Bad and Bcl-xL and inhibiting actions of caspase-9 (Matsuzaki et al. 1999; Pugazhenthi et al. 2000; Tsujimoto and Shimizu 2000). Through this pathway, cell death is inactivated.

Even though most research studied the actions of SNCA in brain diseases, there are evidence showing the ubiquitous presence of alpha-synuclein protein in the gastro-enteric tract and human body fluids such as blood, saliva, and cerebrospinal fluid (Campo et al. 2019; El-Agnaf et al. 2006; Fenyi et al. 2019; Gao et al. 2015). This suggests that the pro-apoptotic action of overexpressed/mutated SNCA can be observed in the peripheral organs too, including those which are prone to cancer development (Gao et al. 2015; Hansson et al. 2014; Wang et al. 2016). Additionally, animal works have shown that α-synuclein can cross the blood-brain barrier due to compromised blood-brain barrier integrity in PD (Brochard et al. 2009; Gray and Woulfe 2015; Peelaerts et al. 2015). In relation to lung cancer, a study by Yan et al. showed that SNCA is downregulated in lung adenocarcinoma (Yan et al. 2018). Bioinformatics analyses done by this group had suggested the tumor suppressive roles of SNCA (Yan et al. 2018). Hence, high expression of SNCA in lung adenocarcinoma patients would improve the overall survival time and post-progression survival time in the lung adenocarcinoma patients (Yan et al. 2018). It is possible that the reduced lung cancer risk in PD may be associated with an increased apoptosis in cancer cells (Figure 1). This could be attributed to the overexpression of SNCA in PD patients, which contributes to a better tumor suppressing ability. On the contrary, downregulation of SNCA in lung cancer patients may have promoted cancer proliferation and inhibition of apoptosis in the cancer cells (Ge and Xu 2016; West et al. 2005).

Figure 1: 
Lung cancer metabolism mediated by PD-associated genes. Elevated expressions of Parkin, PINK1, DJ-1, FBXO7 and UCHL1 and downregulation of SNCA and LRRK2 were detected in lung cancer. These genes regulate cell proliferation, cell survival, apoptosis, mitophagy/autophagy and cell cycle progression via various signaling pathways in order to promote lung carcinogenesis. CDK1/2/6, cyclin-dependent kinase 1/2/6; CIAP1, cellular inhibitor of apoptosis one; DJ-1, parkinsonism associated deglycase; EGFR, epidermal growth factor receptor; Eps 15, epidermal growth factor receptor pathway substrate 15; ERK, extracellular signal-regulated kinase; FBXO7, F-box protein 7; HIF-1α, hypoxia-inducible factor 1α; JNK, c-Jun N-terminal kinases; LRRK2, leucine-rich repeat kinase two; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; PHB2, prohibitin two; PINK1, PTEN-induced putative kinase one; PI3k/Akt/mTOR, phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin; SNCA, alpha synuclein; SIRT1, sirtuin one; TRAF6, tumor necrosis factor receptor-associated factor 6; UCHL1, ubiquitin carboxyl-terminal esterase L1.
Figure 1:

Lung cancer metabolism mediated by PD-associated genes. Elevated expressions of Parkin, PINK1, DJ-1, FBXO7 and UCHL1 and downregulation of SNCA and LRRK2 were detected in lung cancer. These genes regulate cell proliferation, cell survival, apoptosis, mitophagy/autophagy and cell cycle progression via various signaling pathways in order to promote lung carcinogenesis. CDK1/2/6, cyclin-dependent kinase 1/2/6; CIAP1, cellular inhibitor of apoptosis one; DJ-1, parkinsonism associated deglycase; EGFR, epidermal growth factor receptor; Eps 15, epidermal growth factor receptor pathway substrate 15; ERK, extracellular signal-regulated kinase; FBXO7, F-box protein 7; HIF-1α, hypoxia-inducible factor 1α; JNK, c-Jun N-terminal kinases; LRRK2, leucine-rich repeat kinase two; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; PHB2, prohibitin two; PINK1, PTEN-induced putative kinase one; PI3k/Akt/mTOR, phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin; SNCA, alpha synuclein; SIRT1, sirtuin one; TRAF6, tumor necrosis factor receptor-associated factor 6; UCHL1, ubiquitin carboxyl-terminal esterase L1.

A negative association between SNCA and EGFR signaling pathway has also been reported (Yan et al. 2018) (Figure 1). EGFR plays an important role in cells proliferation, differentiation, inflammatory processes and survival (Lemmon and Schlessinger 2010; Schlessinger 2014). Upon binding to a ligand, EGFR undergoes dimerization followed by autophosphorylation that leads to activation of various downstream signaling pathways (Bethune et al. 2010; Guo et al. 2015). In NSCLC, abnormal EGFR trafficking increases cell proliferation via inhibition of apoptosis and thereby induces tumor development (Sigismund et al. 2018). The previous study also suggested that EGFR might suppress the SNCA expression through protein phosphorylation at four possible EGFR phosphorylation sites, which were identified from the full-length SNCA protein sequence (Yan et al. 2018). However, the mechanisms of EGFR on the SNCA regulation in lung adenocarcinoma remain elusive and require more investigations.

Parkin

Parkin gene mutation is the second most common cause of familial PD of which the majority accounts for autosomal recessive PD (Dawson and Dawson 2014; Kitada et al. 1998). Parkin (also known as PARK2) functions as E3 ubiquitin ligase and comprises a ubiquitin-like domain and four zinc-coordinating RING-like domains, including RING0, RING1 IBR and RING2 (Seirafi et al. 2015). It ubiquitinates various cytosolic and outer mitochondrial membrane proteins for the coordination of cellular pathways such as mitochondrial homeostasis, anti-oxidative stress, mitophagy, protein degradation and tumor suppression (Bernardini et al. 2017; Cesari et al. 2003; Pickrell and Youle 2015; Zhang et al. 2016). Under normal circumstances, parkin mediates neuroprotection by activating the NF-κB pathway during stress. Parkin initiates ubiquitination of IKKγ and TRAF2, leading to the upregulation of pro-survival genes (Henn et al. 2007). Phosphorylation of ubiquitin and the recruitment of parkin to the damaged mitochondria by PINK1 result in mitochondrial degradation through mitophagy, which is the autophagy-lysosome pathway, as a way to maintain mitochondrial quality for cell survival (Bogetofte et al. 2019). In PD pathogenesis, parkin mutations lead to the loss of parkin function. Besides, parkin dysfunction due to oxidative stress or dopaminergic stress has been indicated in sporadic PD (LaVoie et al. 2005; Meng et al. 2011). Studies demonstrated that the nonreceptor tyrosine kinase c-Abl phosphorylates parkin, causing the inhibition of parkin’s ubiquitination and the loss of its neuroprotective ability (Imam et al. 2011; Ko et al. 2010). As a result, the loss of parkin activity accumulates the number of depolarized mitochondria that produce reactive oxygen species (ROS) extensively (McWilliams and Muqit 2017; Pickrell and Youle 2015).

A growing number of studies have revealed the tumor suppressive roles of parkin in lung cancer development (Figure 1). According to a data analysis on cBio portal (Cerami et al. 2012), the frequencies of parkin mutations in lung squamous cell carcinoma and lung adenocarcinoma are about 5.6 and 3%, respectively (Xu et al. 2014). A study by Picchio et al. identified the presence of heterogenous deletions of parkin at exon 2 in lung adenocarcinoma cell lines (Cesari et al. 2003; Picchio et al. 2004). The deletions or mutations of parkin inactivate ubiquitination to cyclin E and results in the loss of its tumor suppressive effect in lung cancer cells (Spruck et al. 1999). Cyclin E is an oncogene that is essential to cell cycle progression by regulating the transitions between cell cycle phases (Mazumder et al. 2004). Cyclin E is usually overexpressed in patients with early-stage NSCLC (Müller-Tidow et al. 2001). Notably, parkin loses its ability to ubiquitinate and degrade cyclin E by preventing the binding between cyclin E and CDK2 (Veeriah et al. 2010). This could promote cell cycle progression (Veeriah et al. 2010). Other than that, studies claim that parkin is overexpressed in both NSCLC cell lines and human NSCLC tissue samples (Duan et al. 2019; Park et al. 2019b). Overexpression of parkin in NSCLC was found to inhibit cascades of signaling pathways including cell proliferation, invasion, cell cycle progression as well as apoptosis (Duan et al. 2019). The study by Park et al. also highlighted that depletion of parkin inhibits tumorigenesis via cell cycle arrest at sub G0/G1 phase in NSCLC cells (Park et al. 2019b). In vitro ubiquitination assays showed that parkin deficiency restricts cell cycle progression by inhibiting p21 degradation and cyclin E/CDK2 complex formation (Park et al. 2019b). Moreover, depletion of parkin activates EGFR but suppresses Akt/mTOR pathways and subsequently enhances apoptosis (Duan et al. 2019; Fallon et al. 2006; Husnjak and Dikic 2006; Xiong et al. 2015). Previous study state that parkin protein mediates the monoubiquitination of Eps15, an adaptor protein found in EGFR endocytosis and trafficking (Fallon et al. 2006; Husnjak and Dikic 2006). The authors also revealed the binding of a ubiquitin-like domain in parkin to ubiquitin-interacting motifs of Eps15, which activates EGFR endocytosis and PI3K-Akt signaling pathway, a pathway that promotes cells growth and survival. A study by Zhang et al. revealed that the inner mitochondrial membrane protein called prohibitin 2 (PHB2) is highly expressed in NSCLC cells (Zhang et al. 2020a). PHB2 functions as a mitophagy receptor to the ligand autophagosomal membrane-associated protein LC3 (Wei et al. 2017). PHB2 accumulation upregulates the expressions of mitophagy-associated proteins in the tumor cells which in turn enhances the parkin-mediated mitophagy (Zhang et al. 2020a). In malignant tumors, dysregulated mitophagy is associated with metastasis of cancers and provides a survival advantage to tumor cells (Wang et al. 2020; Zhang et al. 2020a). Cancerous cells eliminate dysfunctional or damaged mitochondria via mitophagy and reduce ROS levels to maintain the cell survival (Humpton et al. 2019; Zhao et al. 2019).

PINK1

PTEN-induced kinase 1 (PINK1), also known as PARK6, is a serine/threonine kinase that contributes to the regulation of mitochondrial quality control pathways via mitophagy (Ge et al. 2020; Gonçalves and Morais 2021; Quinn et al. 2020). The loss-of-function of PINK1 is also associated with autosomal recessive and early onset of PD (Ge et al. 2020). PINK1 and parkin are mutually interdependent in mitophagy activation by which PINK1 is initiated by mitochondrial membrane potential depolarization to phosphorylate ubiquitin and parkin (Gonçalves and Morais 2021). In response to mitochondrial damage, PINK1 recruits parkin from the cytosol to depolarized mitochondria and initiates mitophagy by eliminating the damaged organelles through autophagosome recruitment and ubiquitin proteasomal degradation (McWilliams and Muqit 2017; Pickrell and Youle 2015). In PD, PINK1 mutation is associated with mitochondrial dysfunction, which triggers oxidative stress (Gautier et al. 2008; Matsuda et al. 2013). Likewise, a mutation in PINK1 can lead to failure in recruiting parkin and subsequently result in the accumulation of damaged mitochondria (Gonçalves and Morais 2021). Also, PINK1 deficiency reduces oxidative phosphorylation due to ROS accumulation and transmembrane potential dissipation (Gautier et al. 2008; Gonçalves and Morais 2021; Vara-Perez et al. 2019). This eventually causes neurodegeneration such as loss of dopaminergic neurons of the substantia nigra pars compacta.

Besides, several studies suggested that the putative roles of PINK1 in cancer development involve regulations of mitophagy, cell survival and cell cycle (Dai et al. 2019; Lu et al. 2020; O’Flanagan et al. 2015) (Figure 1). Emerging evidence have reported an upregulation of PINK1 protein expression in both NSCLC cell lines and human NSCLC tissues (Liu et al. 2018; Lu et al. 2020; Zhang et al. 2017). These studies demonstrated that PINK1 promotes cancer cell survival and proliferation which can lead to the emergence of chemoresistance in NSCLC cells via the NF-κB pathway (Chang et al. 2018; Zhang et al. 2017). PINK1 protein binds to tumor necrosis factor receptor-associated factor 6 (TRAF6) and facilitates ubiquitination of TRAF6 whereby it activates the NF-κB pathway (Chen et al. 2011; Zhang et al. 2017). Furthermore, a study by Dai et al. revealed increased ROS production, increased cell death, inhibited mitophagy and reduced cell proliferation in PINK1 depleted NSCLC cell lines (Dai et al. 2019). Researchers also indicated that knockdown of PINK1 enhances apoptosis of lung cancer cells via caspase 9 or caspase 3 activation (Dai et al. 2019; Liu et al. 2018; Zhang et al. 2017). In addition, downregulation of Bcl-2 levels and upregulation of Bax levels have been revealed in lung cancer cells (Liu et al. 2018). Bcl-2 family regulates the apoptotic pathway where Bcl-2, the anti-apoptotic proteins, and Bax, the pro-apoptotic proteins are the two key members (Liu et al. 2018). Hence, high PINK1 expression could serve as an indicator of poor prognosis in NSCLC patients (Chang et al. 2018).

DJ-1

DJ-1 is comprised of 189 amino acids and usually appears as dimers. It is expressed in almost all cells including neurons, glial cells, macrophages and cancerous cells (Ariga et al. 2013). The PARK7 gene encodes DJ-1 and its mutations including L166P, M26I, D149A, A104T, E64D and L10P are linked to familial PD which usually result in loss of function and conformational change of protein (Bonifati et al. 2003; Moore et al. 2005). On top of that, DJ-1 is found to be overexpressed in reactive astrocytes in sporadic PD and other neurodegenerative diseases under oxidative stress (Ariga et al. 2013). DJ-1 is known to be involved in the regulation of transcription factors such as Nrf2, PI3K and p53, mitochondrial regulation, signal transduction and anti-oxidative stress reaction (Ariga et al. 2013; Kim et al. 2005b; McCoy and Cookson 2011; Zhang et al. 2020b). DJ-1 is also a sensor of oxidative stress which protects cells against free radical assault on the three cysteine residues at amino acids 46 (C46), 53 (C53) and 106 (C106) (Taira et al. 2004; Zheng et al. 2018). Of these, C106 of DJ-1 is highly susceptible to oxidative stress and is sequentially oxidized into sulfenated form (–SOH), sulfinated form (–SO2H), and sulfonic form (–SO3H) (Ariga et al. 2013). Oxidation of C106 to SO3H results in inactive form of DJ-1 and lead to the loss of its biological function under extensive oxidative stress (Mita et al. 2018). As a result, excessive oxidized DJ-1 is correlated with the progression of PD due to its loss of function.

On top of that, DJ-1 is responsible for regulation of various transcription factors including nuclear factor Nrf2, PI3K/PKB, and p53 signal pathways upon oxidative stress induction. DJ-1 can either binds to p53 directly to restore the transcriptional activity or stimulate deacylation and suppress p53 transcriptional activity (Dolgacheva et al. 2019). p53 is a tumor suppressor gene that is known as ‘guardian of the genome’ by regulating DNA repair, cell cycle and cell death (Dolgacheva et al. 2019; Feroz and Sheikh 2020). DJ-1 protects neurons against caspase activation and cell death via p53-mediated Bax expression (Bretaud et al. 2007). However, p53 undergoes lysine acetylation to activate cell cycle arrest or cell death pathways and repair damaged DNA in response to damage incurred (Feroz and Sheikh 2020). Increased p53 levels and activities are associated with increased levels of cytokines, caspase 3 and Bax in the brain of the PD patients (Gandhi and Wood 2005; Mogi et al. 2007). DJ-1 can suppress p53 transcriptional activity by decreasing Bax expressions as well as stimulating post-translational modifications of p53 with acetylation and Topors-mediated sumoylation (Dolgacheva et al. 2019; Fan et al. 2008; Shinbo et al. 2005).

Aberrant protein expression of DJ-1 has been implicated in NSCLC and served as a prognostic marker that predicts poor outcomes in lung cancer patients (Kim et al. 2005a; MacKeigan et al. 2003; Zeng et al. 2011). However, the mechanism of DJ-1 in lung cancer remains ambiguous. DJ-1 overexpression is frequently found in various malignancies serving as a potent activator of the PI3K/Akt/mTOR pathway and the inhibitor of p53-mediated apoptosis (Shinbo et al. 2005; Vasseur et al. 2009) (Figure 1). DJ-1 functions as an antagonist of PTEN tumor suppressor to promote cancer cell survival by enhancing Akt phosphorylation (Kim et al. 2005a). Activation of the PI3K-Akt pathway upregulates hypoxia-inducible factor 1α (HIF-1α) expressions under hypoxic conditions leading to a series of cancer signaling events involving angiogenesis, cell proliferation and metastasis (Zhong et al. 1999).

The negative association between DJ-1 and p53 has played a role in lung carcinogenesis. Takahashi-Niki et al. revealed that DJ-1 inhibits p53 activity by activating a lysine deacetylase called SIRT1, the member of Sirtuin family protein, in A549 lung adenocarcinoma cell line (Takahashi-Niki et al. 2016). p53 mutations were detected in 34% of NSCLC patients with a higher frequency in squamous cell carcinomas than adenocarcinomas (Molina-Vila et al. 2014; Zhang et al. 2019). Genetic abnormality of p53 in lung cancers is associated with poor prognosis (Scoccianti et al. 2012; Szymanowska et al. 2005). Evidence also links p53 and Akt to the chemosensitivity against combinational cisplatin chemotherapy in NSCLC (Yilmaz et al. 2019). Cisplatin treatment or cellular stress triggers the phosphorylation of p53 at serine 15 that impairs the EGFR and Akt downstream signaling cascades in NSCLC patients (Loughery et al. 2014; Vasseur et al. 2012). Inactivation of Akt, together with p53 stimulation, contributes to the ROS production. Akt suppresses p53-mediated apoptosis during chemoresistance that activates EGFR to reduce the ROS levels (Zhang et al. 2019). These preliminary findings propose the potential of p53 re-constitution or EGFR inhibitors as the therapeutic options in sensitizing chemoresistant NSCLC cells. In short, DJ-1 is a negative regulator of p53, where the upregulation of DJ-1 promotes lung cancer progression and proliferation by activating the Akt pathway (Jin 2020).

FBXO7

F-box domain-containing protein, FBXO7/PARK15, belongs to the F-box-containing protein (FBP) family that forms ubiquitin ligase complexes with cullin-1 and SKP1. These complexes are involved in regulating proteasomal degradation and cell cycle progression (Ho et al. 2008; Laman 2006; Zhou et al. 2015). At the N-terminus, FBXO7 protein is composed of a ubiquitin-related (UbR) domain, which recruits parkin to regulate mitochondrial quality (Burchell et al. 2013). FBXO7 mutations are genetically associated with autosomal recessive juvenile PD (Fonzo et al. 2009). A previous study has suggested that the FBXO7 protein is a stress response protein (Zhou et al. 2015). FBXO7 mutations (L34R, T22M, R481C, R378G and R498X) in PD can impair the neuroprotective function and facilitate mitochondrial proteotoxicity by stimulating the formation of parkin protein aggregations (Zhou et al. 2015). Ultimately, FBOX7 dysregulation leads to neurodegeneration via impairment of the ubiquitin proteasome system and inhibition of mitophagy (Zhou et al. 2016; Zhou et al. 2015). Wild-type FBXO7 protein plays a vital role in cryoprotection by facilitating mitophagy under stress. However, increased FBXO7 expression leads to aggregation of deleterious FBXO7 proteins in mitochondria which disrupts mitochondria integrity and eventually causes cell death (Zhou et al. 2016).

Emerging evidence have indicated that FBXO7 plays a role in lung tumorigenesis (Laman et al. 2005; Randle and Laman 2016; Wang et al. 2014). Elevation of FBXO7 levels was detected in both lung adenocarcinoma and squamous cell carcinoma. This triggers the cell cycle progression by facilitating the activation of D cyclins and their catalytic subunit, cyclin dependent kinase (CDK6) (Laman et al. 2005; Laman 2006) (Figure 1). Upregulation of FBXO7 is not only inducing the cyclin D-CDK6 complexes formation but also downregulating CDK inhibitors (p21 and p27) expressions (Laman 2006; Meziane et al. 2011). Increased expression of cyclin D-CDK6 complexes enhances the transition of G1 to S phase to accelerate cell proliferation. On top of that, FBXO7 proteins are involved in proteasome-mediated proteolysis of hepatoma upregulated protein (HURP) (Hsu et al. 2004). FBXO7 recruits HURP at its proline-rich domain in a CDK-1-cyclin B phosphorylation-dependent manner. In NSCLC, HURP is reported to be overexpressed in a bioinformatics analysis. The study reported a negative correlation between HURP expression level with overall survival and relapse-free survival (Wang et al. 2018). Additionally, HURP is involved in several cellular processes that induce lung tumor development including cancer proliferation, invasion and migration (Wang et al. 2018). Besides, FBXO7 is found to activate ubiquitination and degradation of cellular inhibitor of apoptosis protein 1 (cIAP1) which leads to reduced NF-κB signaling activity (Chang et al. 2006; Kuiken et al. 2012; Randle and Laman 2016) (Figure 1). As such, FBXO7 is a negative regulator of NF-κB (Randle and Laman 2016). Studies showed that a high level of cIAP1 was reported in human NSCLC tissues, which inhibits apoptosis of the lung cancer cells (Chang et al. 2006; Yang et al. 2016). Thus, high levels of both FBXO7 and cIAP1 disrupt the NF-κB pathway. NF-κB influences lung tumorigenesis by suppressing apoptosis and inducing metastasis and cell proliferation (Rasmi et al. 2020).

UCHL1

Ubiquitin C-terminal hydrolase L1 (UCHL1, also known as PARK5) is a ubiquitin C-terminal hydrolase that is expressed abundantly in neurons (Day and Thompson 2010; Wilkinson et al. 1989). UCHL1 constitutes 1–2% of soluble brain protein and is present in Lewy bodies (Maraganore et al. 2004). UCHL1 functions as a deubiquitinase (DUB) in ubiquitin-dependent proteolysis, a pathway responsible for damaged or misfolded proteins degradation, cell cycle progression and cell death (Ciechanover and Brundin 2003; Spataro et al. 1998). DUBs facilitate tumor suppressive function in a cascade of signaling pathway (Reyes-Turcu et al. 2009). In addition, the dimeric form of UCHL1 acts as a ligase to extend polyubiquitin chains on α-synuclein and tubulin (Bheda et al. 2010; Liu et al. 2002). Although PD pathogenesis mediated by UCHL1 ligase activity remains unclear, Liu et al. hypothesized that UCH1 promotes lysine-63 polyubiquitination and creates an elevation of α-synuclein that interrupting proteasomal degradation (Liu et al. 2002). The study also proposed that lysine-63 polyubiquitination of α-synuclein may promote pathogenic protofibrils formation and lead to neurotoxicity (Liu et al. 2002).

Even though UCHL1 is widely expressed in neurons, several studies have suggested a positive correlation between UCHL1 and lung cancer progression (Liu et al. 2003; Yao et al. 2022). In tumorigenesis, increased deubiquitination of cyclins by UCHL1 is attributed to the uncontrolled growth of somatic cells (Yao et al. 2022) (Figure 1). Increased expression of UCHL1 is detected in the lung cancer cell lines, hence it may be a potential marker for NSCLC and SCLC (Kim et al. 2009; Sasaki et al. 2001; Shimada et al. 2020). A study by Sasaki et al. showed that UCHL1 expressions were closely correlated with T-stages (features of primary tumor) of NSCLCs, but not with N-stages (regional lymph node involvement) (Sasaki et al. 2001). Thus, the results showed that UCHL1 plays a role in the early stage of lung carcinogenesis (Sasaki et al. 2001). Moreover, overexpression of UCH-L1 in lung cancer cells was reported to activate PI3K-Akt and MAPK signalling pathways. This suggests that UCH-L1 overexpression promotes cell survival and tumour progression (Hurst-Kennedy et al. 2012; Kim et al. 2009). Nevertheless, the actions of UCHL1 on lung cancer development and invasion require further investigation to establish a better understanding of its mechanism.

LRRK2

Leucine-rich repeat kinase 2 (LRRK2) gene mutations are one of the leading causes of autosomal dominant PD (Rivero-Ríos et al. 2020; Tolosa et al. 2020). LRRK2 is a large protein (280 kDa) consisting of multiple domains including Ras of Complex (Roc) GTPase, COR (C-terminal of ROC) and other protein–protein interaction domains (Jeong and Lee 2020Rui et al. 2018). Thus, this LRRK2 protein undergoes multifunctional activities by interacting with diverse proteins and exhibiting catalytic activities as GTPase and kinase (Ravinther et al. 2022). LRRK2 is found to be involved in various cellular processes including inflammation, autophagy, cell survival, homeostasis, protein degradation and mitochondrial functions (Funk et al. 2019). There are seven missense LRRK2 mutations namely R1441G, R1441C, R1441H, R1628P, G2019S, G2385R, I2020T and Y1699C found in PD for which G2019S mutation is the most prevalent (Rivero-Ríos et al. 2020; Rui et al. 2018). These mutations lead to gain-of-function mechanisms, where increased LRRK2 kinase phosphorylates Rab proteins at different subcellular localities, thereby affecting downstream pathways and driving towards PD pathogenesis (Berwick et al. 2019). Several studies have revealed that LRRK2 could potentially mediate α-synuclein toxicity by reducing α-synuclein clearance in microglia via endo-lysosomal pathway whereas LRRK2 G2019S enhances α-synuclein aggregation in neurons (Maekawa et al. 2016; Volpicelli-Daley et al. 2016). Besides, LRRK2 is an upstream regulator of mitogen activated protein kinases (MAPK), shown in both in vivo and in vitro studies. LRRK2 phosphorylates apoptosis signal-regulating kinase 1 (ASK1) and several MAPK kinases including p38 MAPK, c-Jun N-terminal kinases (JNKs) and extracellular signal-regulated kinase (ERK) and activates neuronal apoptosis (Ravinther et al. 2022; Yoon et al. 2017). In addition, LRRK2 G2019S mutation enhances the effect of LRRK2 kinase activity by inducing abnormal activation of both MAPK and JNK signaling pathways and ultimately causing neuronal cell death (Rui et al. 2018; Wallings et al. 2015). Furthermore, LRRK2 modulates autophagy in different cell types (Albanese et al. 2019). Several findings showed that LRRK2 regulates autophagy via MEK/ERK pathway in G2019S mutant fibroblasts (Bravo-San Pedro et al. 2013), Beclin-1/PI3K pathway in astrocytic cells (Manzoni et al. 2016), mTOR-dependent pathway in RAW264.7 macrophages or murine BV2 microglial cells (Schapansky et al. 2014) and Ca2+/CaMKK/AMPK pathway in dopaminergic neuroendocrine cells and PC12 cells (Bedford et al. 2016; Gómez-Suaga and Hilfiker 2012). Of these, LRRK2 could possibly initiate another pathway in cancer cells.

A previous study has reported decreased LRRK2 expression in lung adenocarcinoma (Lebovitz et al. 2021). The study has also shown that in vivo LRRK2 knockdown would promote lung tumorigenesis (Lebovitz et al. 2021). As LRRK2 has been implicated in autophagic processes, studies revealed that knockout of LRRK2 could lead to excessive accumulation of proteins that are involved in the impaired induction of autophagy in kidney and lung cells (Herzig et al. 2011; Tong et al. 2010) (Figure 1). The findings revealed abnormal accumulation of secretory lysosomes called lamellar bodies in lung alveolar type II cells of nonhuman primates when LRRK2 production is inhibited (Baptista et al. 2020; Fuji et al. 2015; Herzig et al. 2011). Besides, deficiency of LRRK2 is reported to impair ERK and JNK signalling pathways which contributes to the autophagic dysfunction and cell senescence (Tian et al. 2021). However, the oncogenic mechanism of LRRK2 dysregulation and its effect in lung cancer require more investigation.

Future perspectives

An understanding of the potential roles of these PD-associated genes and their gene products in lung cancer development can be beneficial in strategizing new therapeutic options for lung cancer. For instance, gene therapy can be exploited to regulate the expression profiles of these genes. Gene therapy has been a promising approach to cancer treatment since the emergence of various high-throughput genomic technologies (Belete 2021; Roma-Rodrigues et al. 2020). One of the most widely used techniques is the recombinant DNA technology such as viral vectors, bactofection and non-viral vectors (chemical and physical) that can deliver gene products (Belete 2021). Besides, advancement in CRISPR/Cas9 genome editing technology is now being tested in clinical trials for lung cancer treatment (Nair et al. 2020). In addition, gene-editing technology is explored to combat the drug resistance in various cancers such as breast, colon, and prostate cancers (Domenici et al. 2019; Kawamura et al. 2015; Li et al. 2019). In the future, this might open a new horizon at which the adjuvant uses of gene therapy along with conventional cancer therapy may be obligatory for advanced-stage lung cancer and drug-resistant patients. Other than that, comprehensive molecular profiling of lung cancer can help establishing a precise classification of lung cancer, so it would not rely solely on the morphology and the detection of various molecular markers.

On top of that, the knowledge gained from studying these genes in lung cancer may also help to understand their physiological roles as well as their roles in the pathogenesis of PD. Genes such as LRRK2, despite being identified as a PD-associated gene, are still poorly understood in terms of their biological functions and how they differ in different cell types (Rocha et al. 2022). Therefore, investigations through lung cancer models or biopsied tumor samples might be able to yield insight to close some knowledge gaps and facilitates development of disease modifying strategies for PD treatment.

Conclusions

Although the current epidemiological studies suggest a potential negative association between PD and lung cancer, the molecular mechanisms of the shared genetic determinants between the two diseases in tumorigenesis remain unclear. The cell autonomous and cell non-autonomous gene functions in both diseases must be considered to clarify and resolve the roles of the different genes and their gene products. The evidence listed in this review have shown that proteins encoded by PD-associated genes contribute to mitochondrial dysfunction, cell cycle abnormalities, oxidative stress, cell death and various pathophysiology in both PD and lung cancer. Mutations or loss-of-function of these genes in PD result in neurodegeneration whereas the aberrant expressions of these PD-associated genes in lung cancer cells modulate tumorigenesis. Hence, further investigations of the PD-associated genes in lung cancer are warranted to have a better understanding on their mechanisms of action. This would help in the development of new novel targeted therapeutic strategies for lung cancer patients. Also, an in-depth understanding of these genes and their gene products might be beneficial to the development of biomarkers for early detection of lung cancer.


Corresponding author: Khuen Yen Ng, School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor, Malaysia, E-mail:

Funding source: Fundamental Research Grant Scheme

Award Identifier / Grant number: FRGS/1/2019/SKK08/MUSM/02/2

Funding source: School of Pharmacy Pilot Research Grant

Award Identifier / Grant number: STG-000045

Funding source: High Impact Research Support Fund

Award Identifier / Grant number: STG-000163

About the authors

Yong Qi Leong

Yong Qi Leong is currently in her second year of PhD in Monash University Malaysia. She is awarded with Graduate Research Merit Scholarship offered by Monash University Malaysia. Her research focuses on molecular biology, lung cancer studies and protein–protein interactions. She has four publications throughout her research journey. Also, she is the executive committee of BioMedKL and holds several projects in order to contribute in the community.

Rhun Yian Koh

Dr. Rhun Yian Koh obtained her BSc Biomedical Science (Hons) and PhD (Immunobiology) degrees at the University Putra Malaysia. She is currently a Senior Lecturer of the Division of Applied Biomedical Science, International Medical University. Her research interests include natural product and neuroscience. She has received multiple national and internal research grants for the past few years, and published more than 80 articles. She has also presented her research findings in multiple conferences. She is a member of the Malaysian Biomedical Science Association.

Soi Moi Chye

Associate Professor Dr. Soi Moi Chye graduated from Kaohsiung Medical University, Taiwan, PhD. She has more than 20 years of teaching experience in universities in Taiwan and Malaysia. Her research interests include neuroscience, toxicology, cancer and immunodiagnostic of parasite infection. At present, she has published more than 77 peer-reviewed articles, her RG score is 32.78, and her h-index is 19.

Khuen Yen Ng

Dr. Khuen Yen Ng completed his BSc in Monash University. He then pursued his MSc study in University of New South Wales. Later he undertook his PhD study in Neuroscience in the prestigious Neuroscience Research Australia. His research interest includes: neurodegenerative diseases (including Parksinons’s disease and Alzheimer’s disease), oncology and sleep studies. He is the current advisor of Malaysian Parkinson’s Disease Association and he has published around 50 articles, with h-index of 12.

  1. Author contributions: Khuen Yen Ng conceived the review and Yong Qi Leong drafted the manuscript. Khuen Yen Ng, Rhun Yian Koh and Soi Moi Chye revised and edited the manuscript. Yong Qi Leong created the figure and table with the ideas provided by Khuen Yen Ng. All authors have read and agreed to the published version of the manuscript.

  2. Research funding: This study was supported by the Fundamental Research Grant Scheme (FRGS/1/2019/SKK08/MUSM/02/2) by Ministry of Higher Education Malaysia, School of Pharmacy Pilot Research Grant (STG-000045) and High Impact Research Support Fund (STG-000163) by Monash University Malaysia.

  3. Conflict of interest statement: The authors declare that they have no conflict of interest.

References

Abidin, A.Z., Garassino, M.C., Califano, R., Harle, A., and Blackhall, F. (2010). Targeted therapies in small cell lung cancer: a review. Ther. Adv. Med. Oncol. 2: 25–37, https://doi.org/10.1177/1758834009356014.Search in Google Scholar PubMed PubMed Central

Ahmad, M., Attoub, S., Singh, M.N., Martin, F.L., and El-Agnaf, O.M. (2007). Gamma-synuclein and the progression of cancer. Faseb. J. 21: 3419–3430, https://doi.org/10.1096/fj.07-8379rev.Search in Google Scholar PubMed

Albanese, F., Novello, S., and Morari, M. (2019). Autophagy and LRRK2 in the aging brain. Front. Neurosci. 13: 1352, https://doi.org/10.3389/fnins.2019.01352.Search in Google Scholar PubMed PubMed Central

Allemani, C., Matsuda, T., Di Carlo, V., Harewood, R., Matz, M., Nikšić, M., Bonaventure, A., Valkov, M., Johnson, C.J., Estève, J., et al.. (2018). Global surveillance of trends in cancer survival 2000–14 (CONCORD-3): analysis of individual records for 37 513 025 patients diagnosed with one of 18 cancers from 322 population-based registries in 71 countries. Lancet 391: 1023–1075.Search in Google Scholar

Appel-Cresswell, S., Vilarino-Guell, C., Encarnacion, M., Sherman, H., Yu, I., Shah, B., Weir, D., Thompson, C., Szu-Tu, C., Trinh, J., et al.. (2013). Alpha-synuclein p.H50Q, a novel pathogenic mutation for Parkinson’s disease. Mov. Disord. 28: 811–813, https://doi.org/10.1002/mds.25421.Search in Google Scholar PubMed

Ariga, H., Takahashi-Niki, K., Kato, I., Maita, H., Niki, T., and Iguchi-Ariga, S.M. (2013). Neuroprotective function of DJ-1 in Parkinson’s disease. Oxid. Med. Cell. Longev. 2013: 683920, https://doi.org/10.1155/2013/683920.Search in Google Scholar PubMed PubMed Central

Baptista, M.A.S., Merchant, K., Barrett, T., Bhargava, S., Bryce, D.K., Ellis, J.M., Estrada, A.A., Fell, M.J., Fiske, B.K., Fuji, R.N., et al.. (2020). LRRK2 inhibitors induce reversible changes in nonhuman primate lungs without measurable pulmonary deficits. Sci. Transl. Med. 12: eaav0820, https://doi.org/10.1126/scitranslmed.aav0820.Search in Google Scholar PubMed

Becker, C., Brobert, G.P., Johansson, S., Jick, S.S., and Meier, C.R. (2010). Cancer risk in association with Parkinson disease: a population-based study. Park. Relat. Disord. 16: 186–190, https://doi.org/10.1016/j.parkreldis.2009.11.005.Search in Google Scholar PubMed

Bedford, C., Sears, C., Perez-Carrion, M., Piccoli, G., and Condliffe, S.B. (2016). LRRK2 regulates voltage-gated calcium channel function. Front. Mol. Neurosci. 9: 35, https://doi.org/10.3389/fnmol.2016.00035.Search in Google Scholar PubMed PubMed Central

Belete, T.M. (2021). The current status of gene therapy for the treatment of cancer. Biol. Targets Ther. 15: 67–77, https://doi.org/10.2147/btt.s302095.Search in Google Scholar PubMed PubMed Central

Bernardini, J.P., Lazarou, M., and Dewson, G. (2017). Parkin and mitophagy in cancer. Oncogene 36: 1315–1327, https://doi.org/10.1038/onc.2016.302.Search in Google Scholar PubMed

Berwick, D.C., Heaton, G.R., Azeggagh, S., and Harvey, K. (2019). LRRK2 Biology from structure to dysfunction: research progresses, but the themes remain the same. Mol. Neurodegener. 14: 49, https://doi.org/10.1186/s13024-019-0344-2.Search in Google Scholar PubMed PubMed Central

Bethune, G., Bethune, D., Ridgway, N., and Xu, Z. (2010). Epidermal growth factor receptor (EGFR) in lung cancer: an overview and update. J. Thorac. Dis. 2: 48–51.Search in Google Scholar

Bheda, A., Gullapalli, A., Caplow, M., Pagano, J.S., and Shackelford, J. (2010). Ubiquitin editing enzyme UCH L1 and microtubule dynamics: implication in mitosis. Cell Cycle 9: 980–994, https://doi.org/10.4161/cc.9.5.10934.Search in Google Scholar PubMed PubMed Central

Bogetofte, H., Jensen, P., Ryding, M., Schmidt, S.I., Okarmus, J., Ritter, L., Worm, C.S., Hohnholt, M.C., Azevedo, C., Roybon, L., et al.. (2019). PARK2 mutation causes metabolic disturbances and impaired survival of human iPSC-derived neurons. Front. Cell. Neurosci. 13: 297, https://doi.org/10.3389/fncel.2019.00297.Search in Google Scholar PubMed PubMed Central

Bonifati, V., Rizzu, P., Van Baren, M.J., Schaap, O., Breedveld, G.J., Krieger, E., Dekker, M.C., Squitieri, F., Ibanez, P., Joosse, M., et al.. (2003). Mutations in the DJ-1 gene associated with autosomal recessive early-onset parkinsonism. Science 299: 256–259, https://doi.org/10.1126/science.1077209.Search in Google Scholar PubMed

Bravo-San Pedro, J.M., Niso-Santano, M., Gómez-Sánchez, R., Pizarro-Estrella, E., Aiastui-Pujana, A., Gorostidi, A., Climent, V., López De Maturana, R., Sanchez-Pernaute, R., López De Munain, A., et al.. (2013). The LRRK2 G2019S mutant exacerbates basal autophagy through activation of the MEK/ERK pathway. Cell. Mol. Life Sci. 70: 121–136, https://doi.org/10.1007/s00018-012-1061-y.Search in Google Scholar PubMed

Bray, F., Ferlay, J., Soerjomataram, I., Siegel, R.L., Torre, L.A., and Jemal, A. (2018). Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA A Cancer J. Clin. 68: 394–424, https://doi.org/10.3322/caac.21492.Search in Google Scholar PubMed

Bretaud, S., Allen, C., Ingham, P.W., and Bandmann, O. (2007). p53-dependent neuronal cell death in a DJ-1-deficient zebrafish model of Parkinson’s disease. J. Neurochem. 100: 1626–1635, https://doi.org/10.1111/j.1471-4159.2006.04291.x.Search in Google Scholar PubMed

Brochard, V., Combadiere, B., Prigent, A., Laouar, Y., Perrin, A., Beray-Berthat, V., Bonduelle, O., Alvarez-Fischer, D., Callebert, J., Launay, J.M., et al.. (2009). Infiltration of CD4+ lymphocytes into the brain contributes to neurodegeneration in a mouse model of Parkinson disease. J. Clin. Invest. 119: 182–192.10.1172/JCI36470Search in Google Scholar PubMed PubMed Central

Burchell, V.S., Nelson, D.E., Sanchez-Martinez, A., Delgado-Camprubi, M., Ivatt, R.M., Pogson, J.H., Randle, S.J., Wray, S., Lewis, P.A., Houlden, H., et al.. (2013). The Parkinson’s disease-linked proteins Fbxo7 and Parkin interact to mediate mitophagy. Nat. Neurosci. 16: 1257–1265, https://doi.org/10.1038/nn.3489.Search in Google Scholar PubMed PubMed Central

Butler, B., Sambo, D., and Khoshbouei, H. (2017). Alpha-synuclein modulates dopamine neurotransmission. J. Chem. Neuroanat. 83–84: 41–49, https://doi.org/10.1016/j.jchemneu.2016.06.001.Search in Google Scholar PubMed PubMed Central

Campo, F., Carletti, R., Fusconi, M., Pellicano, C., Pontieri, F.E., Di Gioia, C.R., and De Vincentiis, M. (2019). Alpha-synuclein in salivary gland as biomarker for Parkinson’s disease. Rev. Neurosci. 30: 455–462, https://doi.org/10.1515/revneuro-2018-0064.Search in Google Scholar PubMed

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.. (2012). The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2: 401–404, https://doi.org/10.1158/2159-8290.cd-12-0095.Search in Google Scholar PubMed PubMed Central

Cesari, R., Martin, E.S., Calin, G.A., Pentimalli, F., Bichi, R., Mcadams, H., Trapasso, F., Drusco, A., Shimizu, M., Masciullo, V., et al.. (2003). Parkin, a gene implicated in autosomal recessive juvenile parkinsonism, is a candidate tumor suppressor gene on chromosome 6q25–q27. Proc. Natl. Acad. Sci. U. S. A. 100: 5956–5961, https://doi.org/10.1073/pnas.0931262100.Search in Google Scholar PubMed PubMed Central

Chan, B.A. and Coward, J.I.G. (2013). Chemotherapy advances in small-cell lung cancer. J. Thorac. Dis. 5(Suppl. 5): S565–S578.Search in Google Scholar

Chan, B.A. and Hughes, B.G. (2015). Targeted therapy for non-small cell lung cancer: current standards and the promise of the future. Transl. Lung Cancer Res. 4: 36–54.Search in Google Scholar

Chang, G., Zhang, W., Ma, Y., and Wen, Q. (2018). PINK1 expression is associated with poor prognosis in lung adenocarcinoma. Tohoku J. Exp. Med. 245: 115–121, https://doi.org/10.1620/tjem.245.115.Search in Google Scholar PubMed

Chang, Y.-F., Cheng, C.-M., Chang, L.-K., Jong, Y.-J., and Yuo, C.-Y. (2006). The F-box protein Fbxo7 interacts with human inhibitor of apoptosis protein cIAP1 and promotes cIAP1 ubiquitination. Biochem. Biophys. Res. Commun. 342: 1022–1026, https://doi.org/10.1016/j.bbrc.2006.02.061.Search in Google Scholar PubMed

Chen, J., Jiang, C., and Wang, S. (2013). LDK378: a promising anaplastic lymphoma kinase (ALK) inhibitor. J. Med. Chem. 56: 5673–5674, https://doi.org/10.1021/jm401005u.Search in Google Scholar PubMed

Chen, R., Manochakian, R., James, L., Azzouqa, A.-G., Shi, H., Zhang, Y., Zhao, Y., Zhou, K., and Lou, Y. (2020). Emerging therapeutic agents for advanced non-small cell lung cancer. J. Hematol. Oncol. 13: 58, https://doi.org/10.1186/s13045-020-00881-7.Search in Google Scholar PubMed PubMed Central

Chen, W., Li, Z., Bai, L., and Lin, Y. (2011). NF-kappaB in lung cancer, a carcinogenesis mediator and a prevention and therapy target. Front. Biosci. 16: 1172–1185, https://doi.org/10.2741/3782.Search in Google Scholar PubMed PubMed Central

Ciechanover, A. and Brundin, P. (2003). The ubiquitin proteasome system in neurodegenerative diseases: sometimes the chicken, sometimes the egg. Neuron 40: 427–446, https://doi.org/10.1016/s0896-6273(03)00606-8.Search in Google Scholar PubMed

Conedera, S., Apaydin, H., Li, Y., Yoshino, H., Ikeda, A., Matsushima, T., Funayama, M., Nishioka, K., and Hattori, N. (2016). FBXO7 mutations in Parkinson’s disease and multiple system atrophy. Neurobiol. Aging 40: 192.e1–192.e5, https://doi.org/10.1016/j.neurobiolaging.2016.01.003.Search in Google Scholar PubMed

Cooper, A.A., Gitler, A.D., Cashikar, A., Haynes, C.M., Hill, K.J., Bhullar, B., Liu, K., Xu, K., Strathearn, K.E., Liu, F., et al.. (2006). Alpha-synuclein blocks ER-Golgi traffic and Rab1 rescues neuron loss in Parkinson’s models. Science 313: 324–328, https://doi.org/10.1126/science.1129462.Search in Google Scholar PubMed PubMed Central

Dai, K., Radin, D., and Leonardi, D. (2019). PINK1 depletion sensitizes non-small cell lung cancer to glycolytic inhibitor 3-bromopyruvate: involvement of ROS and mitophagy. Pharmacol. Rep. 71: 1184–1189, https://doi.org/10.1016/j.pharep.2019.08.002.Search in Google Scholar PubMed

Dawson, T.M. and Dawson, V.L. (2014). Parkin plays a role in sporadic Parkinson’s disease. Neurodegener. Dis. 13: 69–71, https://doi.org/10.1159/000354307.Search in Google Scholar PubMed PubMed Central

Day, I.N. and Thompson, R.J. (2010). UCHL1 (PGP 9.5): neuronal biomarker and ubiquitin system protein. Prog. Neurobiol. 90: 327–362, https://doi.org/10.1016/j.pneurobio.2009.10.020.Search in Google Scholar PubMed

Deng, H., Wang, P., and Jankovic, J. (2018). The genetics of Parkinson disease. Ageing Res. Rev. 42: 72–85, https://doi.org/10.1016/j.arr.2017.12.007.Search in Google Scholar PubMed

Doebele, R.C., Pilling, A.B., Aisner, D.L., Kutateladze, T.G., Le, A.T., Weickhardt, A.J., Kondo, K.L., Linderman, D.J., Heasley, L.E., Franklin, W.A., et al.. (2012). Mechanisms of resistance to crizotinib in patients with ALK gene rearranged non-small cell lung cancer. Clin. Cancer Res. 18: 1472–1482, https://doi.org/10.1158/1078-0432.ccr-11-2906.Search in Google Scholar PubMed PubMed Central

Dolgacheva, L.P., Berezhnov, A.V., Fedotova, E.I., Zinchenko, V.P., and Abramov, A.Y. (2019). Role of DJ-1 in the mechanism of pathogenesis of Parkinson’s disease. J. Bioenerg. Biomembr. 51: 175–188, https://doi.org/10.1007/s10863-019-09798-4.Search in Google Scholar PubMed PubMed Central

Domenici, G., Aurrekoetxea-Rodríguez, I., Simões, B.M., Rábano, M., Lee, S.Y., Millán, J.S., Comaills, V., Oliemuller, E., López-Ruiz, J.A., Zabalza, I., et al.. (2019). A Sox2–Sox9 signalling axis maintains human breast luminal progenitor and breast cancer stem cells. Oncogene 38: 3151–3169, https://doi.org/10.1038/s41388-018-0656-7.Search in Google Scholar PubMed PubMed Central

Dómine, M., Moran, T., Isla, D., Martí, J.L., Sullivan, I., Provencio, M., Olmedo, M.E., Ponce, S., Blasco, A., and Cobo, M. (2020). SEOM clinical guidelines for the treatment of small-cell lung cancer (SCLC) (2019). Clin. Transl. Oncol. 22: 245–255, https://doi.org/10.1007/s12094-020-02295-w.Search in Google Scholar PubMed

Duan, H., Lei, Z., Xu, F., Pan, T., Lu, D., Ding, P., Zhu, C., Pan, C., and Zhang, S. (2019). PARK2 suppresses proliferation and tumorigenicity in non-small cell lung cancer. Front. Oncol. 9: 790, https://doi.org/10.3389/fonc.2019.00790.Search in Google Scholar PubMed PubMed Central

Edwards, B.K., Noone, A.M., Mariotto, A.B., Simard, E.P., Boscoe, F.P., Henley, S.J., Jemal, A., Cho, H., Anderson, R.N., Kohler, B.A., et al.. (2014). Annual Report to the Nation on the status of cancer, 1975–2010, featuring prevalence of comorbidity and impact on survival among persons with lung, colorectal, breast, or prostate cancer. Cancer 120: 1290–1314, https://doi.org/10.1002/cncr.28509.Search in Google Scholar PubMed PubMed Central

El-Agnaf, O.M., Salem, S.A., Paleologou, K.E., Curran, M.D., Gibson, M.J., Court, J.A., Schlossmacher, M.G., and Allsop, D. (2006). Detection of oligomeric forms of alpha-synuclein protein in human plasma as a potential biomarker for Parkinson’s disease. FASEB. J. 20: 419–425, https://doi.org/10.1096/fj.03-1449com.Search in Google Scholar PubMed

Fallon, L., Belanger, C.M., Corera, A.T., Kontogiannea, M., Regan-Klapisz, E., Moreau, F., Voortman, J., Haber, M., Rouleau, G., Thorarinsdottir, T., et al.. (2006). A regulated interaction with the UIM protein Eps15 implicates parkin in EGF receptor trafficking and PI(3)K-Akt signalling. Nat. Cell Biol. 8: 834–842, https://doi.org/10.1038/ncb1441.Search in Google Scholar PubMed

Fan, J., Ren, H., Jia, N., Fei, E., Zhou, T., Jiang, P., Wu, M., and Wang, G. (2008). DJ-1 decreases Bax expression through repressing p53 transcriptional activity. J. Biol. Chem. 283: 4022–4030, https://doi.org/10.1074/jbc.m707176200.Search in Google Scholar PubMed

Fenyi, A., Leclair-Visonneau, L., Clairembault, T., Coron, E., Neunlist, M., Melki, R., Derkinderen, P., and Bousset, L. (2019). Detection of alpha-synuclein aggregates in gastrointestinal biopsies by protein misfolding cyclic amplification. Neurobiol. Dis. 129: 38–43, https://doi.org/10.1016/j.nbd.2019.05.002.Search in Google Scholar PubMed

Feroz, W. and Sheikh, A.M.A. (2020). Exploring the multiple roles of guardian of the genome: P53. Egypt. J. Med. Hum. Genet. 21: 49, https://doi.org/10.1186/s43042-020-00089-x.Search in Google Scholar

Fonzo, A.D., Dekker, M.C.J., Montagna, P., Baruzzi, A., Yonova, E.H., Guedes, L.C., Szczerbinska, A., Zhao, T., Dubbel-Hulsman, L.O.M., Wouters, C.H., et al.. (2009). FBXO7 mutations cause autosomal recessive, early-onset parkinsonian-pyramidal syndrome. Neurology 72: 240, https://doi.org/10.1212/01.wnl.0000338144.10967.2b.Search in Google Scholar PubMed

Freedman, D.M., Wu, J., Chen, H., Engels, E.A., Enewold, L.R., Freedman, N.D., Goedert, J.J., Kuncl, R.W., Gail, M.H., and Pfeiffer, R.M. (2016). Associations between cancer and Parkinson’s disease in U.S. elderly adults. Int. J. Epidemiol. 45: 741–751, https://doi.org/10.1093/ije/dyw016.Search in Google Scholar PubMed PubMed Central

Fuji, R.N., Flagella, M., Baca, M., Baptista, S.M.A., Brodbeck, J., Chan, B.K., Fiske, B.K., Honigberg, L., Jubb, A.M., Katavolos, P., et al.. (2015). Effect of selective LRRK2 kinase inhibition on nonhuman primate lung. Sci. Transl. Med. 7: 273ra15, https://doi.org/10.1126/scitranslmed.aaa3634.Search in Google Scholar PubMed

Funk, N., Munz, M., Ott, T., Brockmann, K., Wenninger-Weinzierl, A., Kühn, R., Vogt-Weisenhorn, D., Giesert, F., Wurst, W., Gasser, T., et al.. (2019). The Parkinson’s disease-linked Leucine-rich repeat kinase 2 (LRRK2) is required for insulin-stimulated translocation of GLUT4. Sci. Rep. 9: 4515, https://doi.org/10.1038/s41598-019-40808-y.Search in Google Scholar PubMed PubMed Central

Furrukh, M. (2013). Tobacco smoking and lung cancer: perception-changing facts. Sultan Qaboos Univ. Med. J. 13: 345–358, https://doi.org/10.12816/0003255.Search in Google Scholar PubMed PubMed Central

Gandhi, S. and Wood, N.W. (2005). Molecular pathogenesis of Parkinson’s disease. Hum. Mol. Genet. 14: 2749–2755, https://doi.org/10.1093/hmg/ddi308.Search in Google Scholar PubMed

Gao, L., Tang, H., Nie, K., Wang, L., Zhao, J., Gan, R., Huang, J., Zhu, R., Feng, S., Duan, Z., et al.. (2015). Cerebrospinal fluid alpha-synuclein as a biomarker for Parkinson’s disease diagnosis: a systematic review and meta-analysis. Int. J. Neurosci. 125: 645–654, https://doi.org/10.3109/00207454.2014.961454.Search in Google Scholar PubMed

Garon, E.B., Hellmann, M.D., Rizvi, N.A., Carcereny, E., Leighl, N.B., Ahn, M.-J., Eder, J.P., Balmanoukian, A.S., Aggarwal, C., Horn, L., et al.. (2019). Five-year overall survival for patients with advanced non‒small-cell lung cancer treated with pembrolizumab: results from the phase I KEYNOTE-001 study. J. Clin. Oncol. 37: 2518–2527, https://doi.org/10.1200/jco.19.00934.Search in Google Scholar PubMed PubMed Central

Gautier, C.A., Kitada, T., and Shen, J. (2008). Loss of PINK1 causes mitochondrial functional defects and increased sensitivity to oxidative stress. Proc. Natl. Acad. Sci. U. S. A. 105: 11364–11369, https://doi.org/10.1073/pnas.0802076105.Search in Google Scholar PubMed PubMed Central

Ge, P., Dawson, V.L., and Dawson, T.M. (2020). PINK1 and Parkin mitochondrial quality control: a source of regional vulnerability in Parkinson’s disease. Mol. Neurodegener. 15: 20, https://doi.org/10.1186/s13024-020-00367-7.Search in Google Scholar PubMed PubMed Central

Ge, Y. and Xu, K. (2016). Alpha-synuclein contributes to malignant progression of human meningioma via the Akt/mTOR pathway. Cancer Cell Int. 16: 86, https://doi.org/10.1186/s12935-016-0361-y.Search in Google Scholar PubMed PubMed Central

Gómez-Suaga, P. and Hilfiker, S. (2012). LRRK2 as a modulator of lysosomal calcium homeostasis with downstream effects on autophagy. Autophagy 8: 692–693, https://doi.org/10.4161/auto.19305.Search in Google Scholar PubMed

Gonçalves, F.B. and Morais, V.A. (2021). PINK1: a bridge between mitochondria and Parkinson’s disease. Life 11, https://doi.org/10.3390/life11050371.Search in Google Scholar PubMed PubMed Central

Gray, M.T. and Woulfe, J.M. (2015). Striatal blood-brain barrier permeability in Parkinson’s disease. J. Cerebr. Blood Flow Metabol. 35: 747–750, https://doi.org/10.1038/jcbfm.2015.32.Search in Google Scholar PubMed PubMed Central

Guo, G., Gong, K., Wohlfeld, B., Hatanpaa, K.J., Zhao, D., and Habib, A.A. (2015). Ligand-Independent EGFR signaling. Cancer Res. 75: 3436–3441, https://doi.org/10.1158/0008-5472.can-15-0989.Search in Google Scholar PubMed PubMed Central

Hansson, O., Hall, S., Ohrfelt, A., Zetterberg, H., Blennow, K., Minthon, L., Nagga, K., Londos, E., Varghese, S., Majbour, N.K., et al.. (2014). Levels of cerebrospinal fluid alpha-synuclein oligomers are increased in Parkinson’s disease with dementia and dementia with Lewy bodies compared to Alzheimer’s disease. Alzheimer’s Res. Ther. 6: 25, https://doi.org/10.1186/alzrt255.Search in Google Scholar PubMed PubMed Central

Henn, I.H., Bouman, L., Schlehe, J.S., Schlierf, A., Schramm, J.E., Wegener, E., Nakaso, K., Culmsee, C., Berninger, B., Krappmann, D., et al.. (2007). Parkin mediates neuroprotection through activation of IkappaB kinase/nuclear factor-kappaB signaling. J. Neurosci. 27: 1868–1878, https://doi.org/10.1523/jneurosci.5537-06.2007.Search in Google Scholar PubMed PubMed Central

Herzig, M.C., Kolly, C., Persohn, E., Theil, D., Schweizer, T., Hafner, T., Stemmelen, C., Troxler, T.J., Schmid, P., Danner, S., et al.. (2011). LRRK2 protein levels are determined by kinase function and are crucial for kidney and lung homeostasis in mice. Hum. Mol. Genet. 20: 4209–4223, https://doi.org/10.1093/hmg/ddr348.Search in Google Scholar PubMed PubMed Central

Ho, M., Ou, C., Chan, Y.R., Chien, C.T., and Pi, H. (2008). The utility F-box for protein destruction. Cell. Mol. Life Sci. 65: 1977–2000, https://doi.org/10.1007/s00018-008-7592-6.Search in Google Scholar PubMed

Howlader, N., Noone, A., Krapcho, M., Garshell, J., Miller, D., Altekruse, S., Kosary, C., Yu, M., Ruhl, J., and Tatalovich, Z. (2014). SEER cancer statistics review, 1975–2012. National Cancer Institute, Bethesda, Maryland.Search in Google Scholar

Hsu, J.M., Lee, Y.C., Yu, C.T., and Huang, C.Y. (2004). Fbx7 functions in the SCF complex regulating Cdk1-cyclin B-phosphorylated hepatoma up-regulated protein (HURP) proteolysis by a proline-rich region. J. Biol. Chem. 279: 32592–32602, https://doi.org/10.1074/jbc.m404950200.Search in Google Scholar PubMed

Humpton, T.A.-O., Alagesan, B.A.-O., Denicola, G.A.-O., Lu, D., Yordanov, G.N., Leonhardt, C.S., Yao, M.A., Alagesan, P.A.-O., Zaatari, M.N., Park, Y., et al.. (2019). Oncogenic KRAS induces NIX-mediated mitophagy to promote pancreatic cancer. Cancer Discov. 9: 1268–1287, https://doi.org/10.1158/2159-8290.Search in Google Scholar

Hurst-Kennedy, J., Chin, L.-S., and Li, L. (2012). Ubiquitin C-terminal hydrolase L1 in tumorigenesis. Biochem. Res. Int. 2012: 123706, https://doi.org/10.1155/2012/123706.Search in Google Scholar PubMed PubMed Central

Husnjak, K. and Dikic, I. (2006). EGFR trafficking: parkin’ in a jam. Nat. Cell Biol. 8: 787–788, https://doi.org/10.1038/ncb0806-787.Search in Google Scholar PubMed

Imam, S.Z., Zhou, Q., Yamamoto, A., Valente, A.J., Ali, S.F., Bains, M., Roberts, J.L., Kahle, P.J., Clark, R.A., and Li, S. (2011). Novel regulation of parkin function through c-Abl-mediated tyrosine phosphorylation: implications for Parkinson’s disease. J. Neurosci. 31: 157–163, https://doi.org/10.1523/jneurosci.1833-10.2011.Search in Google Scholar PubMed PubMed Central

Ingelsson, M. (2016). Alpha-synuclein oligomers-neurotoxic molecules in Parkinson’s disease and other Lewy body disorders. Front. Neurosci. 10: 408, https://doi.org/10.3389/fnins.2016.00408.Search in Google Scholar PubMed PubMed Central

Janezic, S., Threlfell, S., Dodson, P.D., Dowie, M.J., Taylor, T.N., Potgieter, D., Parkkinen, L., Senior, S.L., Anwar, S., Ryan, B., et al.. (2013). Deficits in dopaminergic transmission precede neuron loss and dysfunction in a new Parkinson model. Proc. Natl. Acad. Sci. U. S. A. 110: E4016–E4025, https://doi.org/10.1073/pnas.1309143110.Search in Google Scholar PubMed PubMed Central

Jankovic, J. (2008). Parkinson’s disease: clinical features and diagnosis. J. Neurol. Neurosurg. Psychiatry 79: 368–376, https://doi.org/10.1136/jnnp.2007.131045.Search in Google Scholar PubMed

Jeong, G.R. and Lee, B.A.-O. (2020). Pathological functions of LRRK2 in Parkinson’s disease. Cells 9: 2565, https://doi.org/10.3390/cells9122565.Search in Google Scholar PubMed PubMed Central

Jin, W. (2020). Novel insights into PARK7 (DJ-1), a potential anti-cancer therapeutic target, and implications for cancer progression. J. Clin. Med. 9: 1256, https://doi.org/10.3390/jcm9051256.Search in Google Scholar PubMed PubMed Central

Kalia, L.V. and Lang, A.E. (2015). Parkinson’s disease. Lancet 386: 896–912, https://doi.org/10.1016/s0140-6736(14)61393-3.Search in Google Scholar

Kawamura, N., Nimura, K., Nagano, H., Yamaguchi, S., Nonomura, N., and Kaneda, Y. (2015). CRISPR/Cas9-mediated gene knockout of NANOG and NANOGP8 decreases the malignant potential of prostate cancer cells. Oncotarget 6: 22361–22374, https://doi.org/10.18632/oncotarget.4293.Search in Google Scholar PubMed PubMed Central

Kazandjian, D., Blumenthal, G.M., Chen, H.Y., He, K., Patel, M., Justice, R., Keegan, P., and Pazdur, R. (2014). FDA approval summary: crizotinib for the treatment of metastatic non-small cell lung cancer with anaplastic lymphoma kinase rearrangements. Oncology 19: e5–e11, https://doi.org/10.1634/theoncologist.2014-0241.Search in Google Scholar PubMed PubMed Central

Kim, H.C. and Choi, C.M. (2020). Current status of immunotherapy for lung cancer and future perspectives. Tuberc. Respir. Dis. 83: 14–19, https://doi.org/10.4046/trd.2019.0039.Search in Google Scholar PubMed PubMed Central

Kim, H.J., Kim, Y.M., Lim, S., Nam, Y.K., Jeong, J., Kim, H.J., and Lee, K.J. (2009). Ubiquitin C-terminal hydrolase-L1 is a key regulator of tumor cell invasion and metastasis. Oncogene 28: 117–127, https://doi.org/10.1038/onc.2008.364.Search in Google Scholar PubMed

Kim, R.H., Peters, M., Jang, Y., Shi, W., Pintilie, M., Fletcher, G.C., Deluca, C., Liepa, J., Zhou, L., Snow, B., et al.. (2005a). DJ-1, a novel regulator of the tumor suppressor PTEN. Cancer Cell 7: 263–273, https://doi.org/10.1016/j.ccr.2005.02.010.Search in Google Scholar PubMed

Kim, R.H., Smith, P.D., Aleyasin, H., Hayley, S., Mount, M.P., Pownall, S., Wakeham, A., You-Ten, A.J., Kalia, S.K., Horne, P., et al.. (2005b). Hypersensitivity of DJ-1-deficient mice to 1-methyl-4-phenyl-1,2,3, 6-tetrahydropyrindine (MPTP) and oxidative stress. Proc. Natl. Acad. Sci. U. S. A. 102: 5215–5220, https://doi.org/10.1073/pnas.0501282102.Search in Google Scholar PubMed PubMed Central

Kim, W.S., Kagedal, K., and Halliday, G.M. (2014). Alpha-synuclein biology in Lewy body diseases. Alzheimer’s Res. Ther. 6: 73, https://doi.org/10.1186/s13195-014-0073-2.Search in Google Scholar PubMed PubMed Central

Kitada, T., Asakawa, S., Hattori, N., Matsumine, H., Yamamura, Y., Minoshima, S., Yokochi, M., Mizuno, Y., and Shimizu, N. (1998). Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism. Nature 392: 605–608, https://doi.org/10.1038/33416.Search in Google Scholar PubMed

Klein, C. and Westenberger, A. (2012). Genetics of Parkinson’s disease. Cold Spring Harbor Perspect. Med. 2: a008888, https://doi.org/10.1101/cshperspect.a008888.Search in Google Scholar PubMed PubMed Central

Ko, H.S., Lee, Y., Shin, J.H., Karuppagounder, S.S., Gadad, B.S., Koleske, A.J., Pletnikova, O., Troncoso, J.C., Dawson, V.L., and Dawson, T.M. (2010). Phosphorylation by the c-Abl protein tyrosine kinase inhibits parkin’s ubiquitination and protective function. Proc. Natl. Acad. Sci. U. S. A. 107: 16691–16696, https://doi.org/10.1073/pnas.1006083107.Search in Google Scholar PubMed PubMed Central

Kobayashi, S., Boggon, T.J., Dayaram, T., Janne, P.A., Kocher, O., Meyerson, M., Johnson, B.E., Eck, M.J., Tenen, D.G., and Halmos, B. (2005). EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. N. Engl. J. Med. 352: 786–792, https://doi.org/10.1056/nejmoa044238.Search in Google Scholar

Kuiken, H.J., Egan, D.A., Laman, H., Bernards, R., Beijersbergen, R.L., and Dirac, A.M. (2012). Identification of F-box only protein 7 as a negative regulator of NF-kappaB signalling. J. Cell Mol. Med. 16: 2140–2149, https://doi.org/10.1111/j.1582-4934.2012.01524.x.Search in Google Scholar PubMed PubMed Central

Laman, H. (2006). Fbxo7 gets proactive with cyclin D/cdk6. Cell Cycle 5: 279–282, https://doi.org/10.4161/cc.5.3.2403.Search in Google Scholar PubMed

Laman, H., Funes, J.M., Ye, H., Henderson, S., Galinanes-Garcia, L., Hara, E., Knowles, P., Mcdonald, N., and Boshoff, C. (2005). Transforming activity of Fbxo7 is mediated specifically through regulation of cyclin D/cdk6. EMBO J. 24: 3104–3116, https://doi.org/10.1038/sj.emboj.7600775.Search in Google Scholar PubMed PubMed Central

Lavoie, M.J., Ostaszewski, B.L., Weihofen, A., Schlossmacher, M.G., and Selkoe, D.J. (2005). Dopamine covalently modifies and functionally inactivates parkin. Nat. Med. 11: 1214–1221, https://doi.org/10.1038/nm1314.Search in Google Scholar PubMed

Lebovitz, C., Wretham, N., Osooly, M., Milne, K., Dash, T., Thornton, S., Tessier-Cloutier, B., Sathiyaseelan, P., Bortnik, S., Go, N.E., et al.. (2021). Loss of Parkinson’s susceptibility gene LRRK2 promotes carcinogen-induced lung tumorigenesis. Sci. Rep. 11: 2097, https://doi.org/10.1038/s41598-021-81639-0.Search in Google Scholar PubMed PubMed Central

Lemmon, M.A. and Schlessinger, J. (2010). Cell signaling by receptor tyrosine kinases. Cell 141: 1117–1134, https://doi.org/10.1016/j.cell.2010.06.011.Search in Google Scholar PubMed PubMed Central

Leong, Y.Q., Lee, S.W.H., and Ng, K.Y. (2021). Cancer risk in Parkinson disease: an updated systematic review and meta-analysis. Eur. J. Neurol. 28: 4219–4237, https://doi.org/10.1111/ene.15069.Search in Google Scholar PubMed

Lesage, S., Anheim, M., Letournel, F., Bousset, L., Honore, A., Rozas, N., Pieri, L., Madiona, K., Durr, A., Melki, R., et al.. (2013). G51D alpha-synuclein mutation causes a novel parkinsonian-pyramidal syndrome. Ann. Neurol. 73: 459–471, https://doi.org/10.1002/ana.23894.Search in Google Scholar PubMed

Li, A.R., Chitale, D., Riely, G.J., Pao, W., Miller, V.A., Zakowski, M.F., Rusch, V., Kris, M.G., and Ladanyi, M. (2008). EGFR mutations in lung adenocarcinomas: clinical testing experience and relationship to EGFR gene copy number and immunohistochemical expression. J. Mol. Diagn. 10: 242–248, https://doi.org/10.2353/jmoldx.2008.070178.Search in Google Scholar PubMed PubMed Central

Li, W., Cho, M.-Y., Lee, S., Jang, M., Park, J., and Park, R. (2019). CRISPR-Cas9 mediated CD133 knockout inhibits colon cancer invasion through reduced epithelial-mesenchymal transition. PLoS One 14: e0220860, https://doi.org/10.1371/journal.pone.0220860.Search in Google Scholar PubMed PubMed Central

Liu, L., Zuo, Z., Lu, S., Wang, L., Liu, A., and Liu, X. (2018). Silencing of PINK1 represses cell growth, migration and induces apoptosis of lung cancer cells. Biomed. Pharmacother. 106: 333–341, https://doi.org/10.1016/j.biopha.2018.06.128.Search in Google Scholar PubMed

Liu, Q.X., Zheng, H., Deng, X.F., Zhou, D., and Dai, J.G. (2015). Status of the Parkinson’s disease gene family expression in non-small-cell lung cancer. World J. Surg. Oncol. 13: 238, https://doi.org/10.1186/s12957-015-0646-y.Search in Google Scholar PubMed PubMed Central

Liu, Y., Fallon, L., Lashuel, H.A., Liu, Z., and Lansbury, P.T.Jr. (2002). The UCH-L1 gene encodes two opposing enzymatic activities that affect alpha-synuclein degradation and Parkinson’s disease susceptibility. Cell 111: 209–218, https://doi.org/10.1016/s0092-8674(02)01012-7.Search in Google Scholar PubMed

Liu, Y., Lashuel, H.A., Choi, S., Xing, X., Case, A., Ni, J., Yeh, L.-A., Cuny, G.D., Stein, R.L., and Lansbury, P.T.Jr. (2003). Discovery of inhibitors that elucidate the role of UCH-L1 activity in the H1299 lung cancer cell line. Cell Chem. Biol. 10: 837–846, https://doi.org/10.1016/j.chembiol.2003.08.010.Search in Google Scholar PubMed

Loughery, J., Cox, M., Smith, L.M., and Meek, D.W. (2014). Critical role for p53-serine 15 phosphorylation in stimulating transactivation at p53-responsive promoters. Nucleic Acids Res. 42: 7666–7680, https://doi.org/10.1093/nar/gku501.Search in Google Scholar PubMed PubMed Central

Lu, X., Liu, Q.-X., Zhang, J., Zhou, D., Yang, G.-X., Li, M.-Y., Qiu, Y., Chen, Q., Zheng, H., and Dai, J.-G. (2020). PINK1 overexpression promotes cell migration and proliferation via regulation of autophagy and predicts a poor prognosis in lung cancer cases. Cancer Manag. Res. 12: 7703–7714, https://doi.org/10.2147/cmar.s262466.Search in Google Scholar PubMed PubMed Central

Mackeigan, J.P., Clements, C.M., Lich, J.D., Pope, R.M., Hod, Y., and Ting, J.P.Y. (2003). Proteomic profiling drug-induced apoptosis in non-small cell lung carcinoma. Cancer Res. 63: 6928.Search in Google Scholar

Maekawa, T., Sasaoka, T., Azuma, S., Ichikawa, T., Melrose, H.L., Farrer, M.J., and Obata, F. (2016). Leucine-rich repeat kinase 2 (LRRK2) regulates α-synuclein clearance in microglia. BMC Neurosci. 17: 77, https://doi.org/10.1186/s12868-016-0315-2.Search in Google Scholar PubMed PubMed Central

Maiti, P., Manna, J., and Dunbar, G.L. (2017). Current understanding of the molecular mechanisms in Parkinson’s disease: targets for potential treatments. Transl. Neurodegener. 6: 28, https://doi.org/10.1186/s40035-017-0099-z.Search in Google Scholar PubMed PubMed Central

Majeed, U., Manochakian, R., Zhao, Y., and Lou, Y. (2021). Targeted therapy in advanced non-small cell lung cancer: current advances and future trends. J. Hematol. Oncol. 14: 108, https://doi.org/10.1186/s13045-021-01121-2.Search in Google Scholar PubMed PubMed Central

Manzoni, C., Mamais, A., Roosen, D.A., Dihanich, S., Soutar, M.P.M., Plun-Favreau, H., Bandopadhyay, R., Hardy, J., Tooze, S.A., Cookson, M.R., et al.. (2016). mTOR independent regulation of macroautophagy by leucine rich repeat kinase 2 via beclin-1. Sci. Rep. 6: 35106, https://doi.org/10.1038/srep35106.Search in Google Scholar PubMed PubMed Central

Maraganore, D.M., Lesnick, T.G., Elbaz, A., Chartier-Harlin, M.C., Gasser, T., Kruger, R., Hattori, N., Mellick, G.D., Quattrone, A., Satoh, J., et al.. (2004). UCHL1 is a Parkinson’s disease susceptibility gene. Ann. Neurol. 55: 512–521, https://doi.org/10.1002/ana.20017.Search in Google Scholar PubMed

Masters, G.A., Temin, S., Azzoli, C.G., Giaccone, G., Baker, S.Jr., Brahmer, J.R., Ellis, P.M., Gajra, A., Rackear, N., Schiller, J.H., et al.. (2015). Systemic therapy for stage IV non-small-cell lung cancer: American society of clinical oncology clinical practice guideline update. J. Clin. Oncol. 33: 3488–3515, https://doi.org/10.1200/jco.2015.62.1342.Search in Google Scholar PubMed PubMed Central

Matsuda, S., Kitagishi, Y., and Kobayashi, M. (2013). Function and characteristics of PINK1 in mitochondria. Oxid. Med. Cell. Longev. 2013: 601587, https://doi.org/10.1155/2013/601587.Search in Google Scholar PubMed PubMed Central

Matsuzaki, H., Tamatani, M., Mitsuda, N., Namikawa, K., Kiyama, H., Miyake, S., and Tohyama, M. (1999). Activation of Akt kinase inhibits apoptosis and changes in Bcl-2 and Bax expression induced by nitric oxide in primary hippocampal neurons. J. Neurochem. 73: 2037–2046.10.1046/j.1471-4159.1999.02037.xSearch in Google Scholar

Mazumder, S., Dupree, E.L., and Almasan, A. (2004). A dual role of cyclin E in cell proliferation and apoptosis may provide a target for cancer therapy. Curr. Cancer Drug Targets 4: 65–75, https://doi.org/10.2174/1568009043481669.Search in Google Scholar PubMed PubMed Central

Mccoy, M.K. and Cookson, M.R. (2011). DJ-1 regulation of mitochondrial function and autophagy through oxidative stress. Autophagy 7: 531–532, https://doi.org/10.4161/auto.7.5.14684.Search in Google Scholar PubMed PubMed Central

Mcwilliams, T.G. and Muqit, M.M.K. (2017). PINK1 and Parkin: emerging themes in mitochondrial homeostasis. Curr. Opin. Cell Biol. 45: 83–91, https://doi.org/10.1016/j.ceb.2017.03.013.Search in Google Scholar PubMed

Meade, R.M., Fairlie, D.P., and Mason, J.M. (2019). Alpha-synuclein structure and Parkinson’s disease – lessons and emerging principles. Mol. Neurodegener. 14: 29, https://doi.org/10.1186/s13024-019-0329-1.Search in Google Scholar PubMed PubMed Central

Meng, F., Yao, D., Shi, Y., Kabakoff, J., Wu, W., Reicher, J., Ma, Y., Moosmann, B., Masliah, E., Lipton, S.A., et al.. (2011). Oxidation of the cysteine-rich regions of parkin perturbs its E3 ligase activity and contributes to protein aggregation. Mol. Neurodegener. 6: 34, https://doi.org/10.1186/1750-1326-6-34.Search in Google Scholar PubMed PubMed Central

Meziane, E.K., Randle, S.J., Nelson, D.E., Lomonosov, M., and Laman, H. (2011). Knockdown of Fbxo7 reveals its regulatory role in proliferation and differentiation of haematopoietic precursor cells. J. Cell Sci. 124: 2175, https://doi.org/10.1242/jcs.080465.Search in Google Scholar PubMed

Mita, Y., Kataoka, Y., Saito, Y., Kashi, T., Hayashi, K., Iwasaki, A., Imanishi, T., Miyasaka, T., and Noguchi, N. (2018). Distribution of oxidized DJ-1 in Parkinson’s disease-related sites in the brain and in the peripheral tissues: effects of aging and a neurotoxin. Sci. Rep. 8: 12056, https://doi.org/10.1038/s41598-018-30561-z.Search in Google Scholar PubMed PubMed Central

Mogi, M., Kondo, T., Mizuno, Y., and Nagatsu, T. (2007). p53 protein, interferon-γ, and NF-κB levels are elevated in the parkinsonian brain. Neurosci. Lett. 414: 94–97, https://doi.org/10.1016/j.neulet.2006.12.003.Search in Google Scholar PubMed

Mok, T.S., Wu, Y.L., Ahn, M.J., Garassino, M.C., Kim, H.R., Ramalingam, S.S., Shepherd, F.A., He, Y., Akamatsu, H., Theelen, W.S., et al.. (2017). Osimertinib or platinum-pemetrexed in EGFR T790M-positive lung cancer. N. Engl. J. Med. 376: 629–640, https://doi.org/10.1056/nejmoa1612674.Search in Google Scholar PubMed PubMed Central

Molina-Vila, M.A., Bertran-Alamillo, J., Gascó, A., Mayo-De-Las-Casas, C., Sánchez-Ronco, M., Pujantell-Pastor, L., Bonanno, L., Favaretto, A.G., Cardona, A.F., Vergnenègre, A., et al.. (2014). Nondisruptive p53 mutations are associated with shorter survival in patients with advanced non–small cell lung cancer. Clin. Cancer Res. 20: 4647, https://doi.org/10.1158/1078-0432.ccr-13-2391.Search in Google Scholar PubMed

Moore, D.J., West, A.B., Dawson, V.L., and Dawson, T.M. (2005). Molecular pathophysiology of Parkinson’s disease. Annu. Rev. Neurosci. 28: 57–87, https://doi.org/10.1146/annurev.neuro.28.061604.135718.Search in Google Scholar PubMed

Müller-Tidow, C., Metzger, R., Kügler, K., Diederichs, S., Idos, G., Thomas, M., Dockhorn-Dworniczak, B., Schneider, P.M., Koeffler, H.P., Berdel, W.E., et al.. (2001). Cyclin E is the only cyclin-dependent kinase 2-associated cyclin that predicts metastasis and survival in early stage non-small cell lung cancer. Cancer Res. 61: 647.Search in Google Scholar

Nair, J., Nair, A., Veerappan, S., and Sen, D. (2020). Translatable gene therapy for lung cancer using Crispr CAS9 – an exploratory review. Cancer Gene Ther. 27: 116–124, https://doi.org/10.1038/s41417-019-0116-8.Search in Google Scholar PubMed

O’flanagan, C.H., Morais, V.A., Wurst, W., De Strooper, B., and O’neill, C. (2015). The Parkinson’s gene PINK1 regulates cell cycle progression and promotes cancer-associated phenotypes. Oncogene 34: 1363–1374, https://doi.org/10.1038/onc.2014.81.Search in Google Scholar PubMed

Oczkowska, A., Kozubski, W., Lianeri, M., and Dorszewska, J. (2013). Mutations in PRKN and SNCA genes important for the progress of Parkinson’s disease. Curr. Genom. 14: 502–517, https://doi.org/10.2174/1389202914666131210205839.Search in Google Scholar PubMed PubMed Central

Onoi, K., Chihara, Y., Uchino, J., Shimamoto, T., Morimoto, Y., Iwasaku, M., Kaneko, Y., Yamada, T., and Takayama, K. (2020). Immune checkpoint inhibitors for lung cancer treatment: a review. J. Clin. Med. 9: 1362, https://doi.org/10.3390/jcm9051362.Search in Google Scholar PubMed PubMed Central

Papadimitrakopoulou, V.A., Wu, Y.L., Han, J.Y., Ahn, M.J., Ramalingam, S.S., John, T., Okamoto, I., Yang, J.C.H., Bulusu, K.C., Laus, G., et al.. (2018). Analysis of resistance mechanisms to osimertinib in patients with EGFR T790M advanced NSCLC from the AURA3 study. Ann. Oncol. 29: viii741, https://doi.org/10.1093/annonc/mdy424.064.Search in Google Scholar

Park, J.H., Kim, D.H., Park, Y.G., Kwon, D.Y., Choi, M., Jung, J.H., and Han, K. (2019a). Cancer risk in patients with Parkinson’s disease in South Korea: a nationwide, population-based cohort study. Eur. J. Cancer 117: 5–13, https://doi.org/10.1016/j.ejca.2019.04.033.Search in Google Scholar PubMed

Park, K.R., Yun, J.S., Park, M.H., Jung, Y.Y., Yeo, I.J., Nam, K.T., Kim, H.D., Song, J.K., Choi, D.Y., Park, P.H., et al.. (2019b). Loss of parkin reduces lung tumor development by blocking p21 degradation. PLoS One 14: e0217037, https://doi.org/10.1371/journal.pone.0217037.Search in Google Scholar PubMed PubMed Central

Peelaerts, W., Bousset, L., Van Der Perren, A., Moskalyuk, A., Pulizzi, R., Giugliano, M., Van Den Haute, C., Melki, R., and Baekelandt, V. (2015). α-Synuclein strains cause distinct synucleinopathies after local and systemic administration. Nature 522: 340–344, https://doi.org/10.1038/nature14547.Search in Google Scholar PubMed

Picchio, M.C., Martin, E.S., Cesari, R., Calin, G.A., Yendamuri, S., Kuroki, T., Pentimalli, F., Sarti, M., Yoder, K., Kaiser, L.R., et al.. (2004). Alterations of the tumor suppressor gene in non-small cell lung cancer. Clin. Cancer Res. 10: 2720–2724, https://doi.org/10.1158/1078-0432.ccr-03-0086.Search in Google Scholar PubMed

Pickrell, A.M. and Youle, R.J. (2015). The roles of PINK1, parkin, and mitochondrial fidelity in Parkinson’s disease. Neuron 85: 257–273, https://doi.org/10.1016/j.neuron.2014.12.007.Search in Google Scholar PubMed PubMed Central

Pikor, L.A., Ramnarine, V.R., Lam, S., and Lam, W.L. (2013). Genetic alterations defining NSCLC subtypes and their therapeutic implications. Lung Cancer 82: 179–189, https://doi.org/10.1016/j.lungcan.2013.07.025.Search in Google Scholar PubMed

Postuma, R.B., Berg, D., Stern, M., Poewe, W., Olanow, C.W., Oertel, W., Obeso, J., Marek, K., Litvan, I., Lang, A.E., et al.. (2015). MDS clinical diagnostic criteria for Parkinson’s disease. Mov. Disord. 30: 1591–1601, https://doi.org/10.1002/mds.26424.Search in Google Scholar PubMed

Proukakis, C., Dudzik, C.G., Brier, T., Mackay, D.S., Cooper, J.M., Millhauser, G.L., Houlden, H., and Schapira, A.H. (2013). A novel alpha-synuclein missense mutation in Parkinson disease. Neurology 80: 1062–1064, https://doi.org/10.1212/wnl.0b013e31828727ba.Search in Google Scholar

Pugazhenthi, S., Nesterova, A., Sable, C., Heidenreich, K.A., Boxer, L.M., Heasley, L.E., and Reusch, J.E. (2000). Akt/protein kinase B up-regulates Bcl-2 expression through cAMP-response element-binding protein. J. Biol. Chem. 275: 10761–10766, https://doi.org/10.1074/jbc.275.15.10761.Search in Google Scholar PubMed

Puzanov, I., Diab, A., Abdallah, K., Bingham, C.O.3rd, Brogdon, C., Dadu, R., Hamad, L., Kim, S., Lacouture, M.E., Leboeuf, N.R., et al.. (2017). Managing toxicities associated with immune checkpoint inhibitors: consensus recommendations from the society for immunotherapy of cancer (SITC) toxicity management working group. J. Immunother. Cancer 5: 95, https://doi.org/10.1186/s40425-017-0300-z.Search in Google Scholar PubMed PubMed Central

Quinn, P.M.J., Moreira, P.I., Ambrósio, A.F., and Alves, C.H. (2020). PINK1/PARKIN signalling in neurodegeneration and neuroinflammation. Acta Neuropathol. Commun. 8: 189, https://doi.org/10.1186/s40478-020-01062-w.Search in Google Scholar PubMed PubMed Central

Ragland, M., Hutter, C., Zabetian, C., and Edwards, K. (2009). Association between the ubiquitin carboxyl-terminal esterase L1 gene (UCHL1) S18Y variant and Parkinson’s Disease: a HuGE review and meta-analysis. Am. J. Epidemiol. 170: 1344–1357, https://doi.org/10.1093/aje/kwp288.Search in Google Scholar PubMed PubMed Central

Raman, V., Yang, C.-F.J., Deng, J.Z., and D’amico, T.A. (2018). Surgical treatment for early stage non-small cell lung cancer. J. Thorac. Dis. 10: S898–S904, https://doi.org/10.21037/jtd.2018.01.172.Search in Google Scholar PubMed PubMed Central

Randle, S.J. and Laman, H. (2016). F-box protein interactions with the hallmark pathways in cancer. Semin. Cancer Biol. 36: 3–17, https://doi.org/10.1016/j.semcancer.2015.09.013.Search in Google Scholar PubMed

Rasmi, R.R., Sakthivel, K.M., and Guruvayoorappan, C. (2020). NF-κB inhibitors in treatment and prevention of lung cancer. Biomed. Pharmacother. 130: 110569, https://doi.org/10.1016/j.biopha.2020.110569.Search in Google Scholar PubMed

Ravinther, A.I., Dewadas, H.D., Tong, S.R., Foo, C.N., Lin, Y.-E., Chien, C.-T., and Lim, Y.M. (2022). Molecular pathways involved in LRRK2-linked Parkinsonandrsquo;s disease: a systematic review. Int. J. Mol. Sci. 23: 11744, https://doi.org/10.3390/ijms231911744.Search in Google Scholar PubMed PubMed Central

Reyes-Turcu, F.E., Ventii, K.H., and Wilkinson, K.D. (2009). Regulation and cellular roles of ubiquitin-specific deubiquitinating enzymes. Annu. Rev. Biochem. 78: 363–397, https://doi.org/10.1146/annurev.biochem.78.082307.091526.Search in Google Scholar PubMed PubMed Central

Richer, A.L., Friel, J.M., Carson, V.M., Inge, L.J., and Whitsett, T.G. (2015). Genomic profiling toward precision medicine in non-small cell lung cancer: getting beyond EGFR. Pharmgenomics Pers. Med. 8: 63–79, https://doi.org/10.2147/pgpm.s52845.Search in Google Scholar

Rivero-Ríos, P., Romo-Lozano, M., Fasiczka, R., Naaldijk, Y., and Hilfiker, S. (2020). LRRK2-Related Parkinson’s disease due to altered endolysosomal biology with variable Lewy body pathology: a hypothesis. Front. Neurosci. 14: 556, https://doi.org/10.3389/fnins.2020.00556.Search in Google Scholar PubMed PubMed Central

Rocha, E.M., Keeney, M.T., Di Maio, R., De Miranda, B.R., and Greenamyre, J.T. (2022). LRRK2 and idiopathic Parkinson’s disease. Trends Neurosci. 45: 224–236, https://doi.org/10.1016/j.tins.2021.12.002.Search in Google Scholar PubMed PubMed Central

Roma-Rodrigues, C., Rivas-García, L., Baptista, P.V., and Fernandes, A.R. (2020). Gene therapy in cancer treatment: why go nano? Pharmaceutics 12: 233, https://doi.org/10.3390/pharmaceutics12030233.Search in Google Scholar PubMed PubMed Central

Rothschild, S.I. (2014). Ceritinib-a second-generation ALK inhibitor overcoming resistance in ALK-rearranged non-small cell lung cancer. Transl. Lung Cancer Res. 3: 379–381.Search in Google Scholar

Rui, Q., Ni, H., Li, D., Gao, R., and Chen, G. (2018). The role of LRRK2 in neurodegeneration of Parkinson disease. Curr. Neuropharmacol. 16: 1348–1357, https://doi.org/10.2174/1570159x16666180222165418.Search in Google Scholar

Saltos, A., Shafique, M., and Chiappori, A. (2020). Update on the biology, management, and treatment of small cell lung cancer (SCLC). Front. Oncol. 10: 1074, https://doi.org/10.3389/fonc.2020.01074.Search in Google Scholar PubMed PubMed Central

Sasaki, H., Yukiue, H., Moriyama, S., Kobayashi, Y., Nakashima, Y., Kaji, M., Fukai, I., Kiriyama, M., Yamakawa, Y., and Fujii, Y. (2001). Expression of the protein gene product 9.5, PGP9.5, is correlated with T-status in non-small cell lung cancer. Jpn. J. Clin. Oncol. 31: 532–535, https://doi.org/10.1093/jjco/hye113.Search in Google Scholar PubMed

Schad, F., Thronicke, A., Steele, M.L., Merkle, A., Matthes, B., Grah, C., and Matthes, H. (2018). Overall survival of stage IV non-small cell lung cancer patients treated with Viscum album L. in addition to chemotherapy, a real-world observational multicenter analysis. PLoS One 13: e0203058, https://doi.org/10.1371/journal.pone.0203058.Search in Google Scholar PubMed PubMed Central

Schapansky, J., Nardozzi, J.D., Felizia, F., and Lavoie, M.J. (2014). Membrane recruitment of endogenous LRRK2 precedes its potent regulation of autophagy. Hum. Mol. Genet. 23: 4201–4214, https://doi.org/10.1093/hmg/ddu138.Search in Google Scholar PubMed PubMed Central

Schlessinger, J. (2014). Receptor tyrosine kinases: legacy of the first two decades. Cold Spring Harbor Perspect. Biol. 6: a008912, https://doi.org/10.1101/cshperspect.a008912.Search in Google Scholar PubMed PubMed Central

Scoccianti, C., Vesin, A., Martel, G., Olivier, M., Brambilla, E., Timsit, J.-F., Tavecchio, L., Brambilla, C., Field, J.K., and Hainaut, P. (2012). Prognostic value of TP53, KRAS and EGFR mutations in nonsmall cell lung cancer: the EUELC cohort. Eur. Respir. J. 40: 177, https://doi.org/10.1183/09031936.00097311.Search in Google Scholar PubMed

Seirafi, M., Kozlov, G., and Gehring, K. (2015). Parkin structure and function. FEBS J. 282: 2076–2088, https://doi.org/10.1111/febs.13249.Search in Google Scholar PubMed PubMed Central

Selvaraj, S. and Piramanayagam, S. (2019). Impact of gene mutation in the development of Parkinson’s disease. Gene Dis. 6: 120–128, https://doi.org/10.1016/j.gendis.2019.01.004.Search in Google Scholar PubMed PubMed Central

Seo, J.H., Rah, J.C., Choi, S.H., Shin, J.K., Min, K., Kim, H.S., Park, C.H., Kim, S., Kim, E.M., Lee, S.H., et al.. (2002). Alpha-synuclein regulates neuronal survival via Bcl-2 family expression and PI3/Akt kinase pathway. Faseb. J. 16: 1826–1828.10.1096/fj.02-0041fjeSearch in Google Scholar PubMed

Shimada, Y., Kudo, Y., Maehara, S., Matsubayashi, J., Otaki, Y., Kajiwara, N., Ohira, T., Minna, J.D., and Ikeda, N. (2020). Ubiquitin C-terminal hydrolase-L1 has prognostic relevance and is a therapeutic target for high-grade neuroendocrine lung cancers. Cancer Sci. 111: 610–620, https://doi.org/10.1111/cas.14284.Search in Google Scholar PubMed PubMed Central

Shinbo, Y., Taira, T., Niki, T., Iguchi-Ariga, S.M., and Ariga, H. (2005). DJ-1 restores p53 transcription activity inhibited by Topors/p53BP3. Int. J. Oncol. 26: 641–648, https://doi.org/10.3892/ijo.26.3.641.Search in Google Scholar

Sigismund, S., Avanzato, D., and Lanzetti, L. (2018). Emerging functions of the EGFR in cancer. Mol. Oncol. 12: 3–20, https://doi.org/10.1002/1878-0261.12155.Search in Google Scholar PubMed PubMed Central

Snead, D. and Eliezer, D. (2014). Alpha-synuclein function and dysfunction on cellular membranes. Exp. Neurobiol. 23: 292–313, https://doi.org/10.5607/en.2014.23.4.292.Search in Google Scholar PubMed PubMed Central

Solomon, B.J., Mok, T., Kim, D.-W., Wu, Y.-L., Nakagawa, K., Mekhail, T., Felip, E., Cappuzzo, F., Paolini, J., Usari, T., et al.. (2014). First-line crizotinib versus chemotherapy in ALK-positive lung cancer. N. Engl. J. Med. 371: 2167–2177, https://doi.org/10.1056/nejmoa1408440.Search in Google Scholar

Soria, J.C., Ohe, Y., Vansteenkiste, J., Reungwetwattana, T., Chewaskulyong, B., Lee, K.H., Dechaphunkul, A., Imamura, F., Nogami, N., Kurata, T., et al.. (2018). Osimertinib in untreated EGFR-mutated advanced non-small-cell lung cancer. N. Engl. J. Med. 378: 113–125, https://doi.org/10.1056/nejmoa1713137.Search in Google Scholar PubMed

Spataro, V., Norbury, C., and Harris, A.L. (1998). The ubiquitin-proteasome pathway in cancer. Br. J. Cancer 77: 448–455, https://doi.org/10.1038/bjc.1998.71.Search in Google Scholar PubMed PubMed Central

Spillantini, M.G. (1999). Parkinson’s disease, dementia with Lewy bodies and multiple system atrophy are alpha-synucleinopathies. Park. Relat. Disord. 5: 157–162, https://doi.org/10.1016/s1353-8020(99)00031-0.Search in Google Scholar PubMed

Spruck, C.H., Won, K.A., and Reed, S.I. (1999). Deregulated cyclin E induces chromosome instability. Nature 401: 297–300, https://doi.org/10.1038/45836.Search in Google Scholar PubMed

Stefanis, L. (2012). α-Synuclein in Parkinson’s disease. Cold Spring Harbor Perspect. Med. 2: a009399, https://doi.org/10.1101/cshperspect.a009399.Search in Google Scholar PubMed PubMed Central

Sun, J.M., Ahn, M.J., Choi, Y.L., Ahn, J.S., and Park, K. (2013). Clinical implications of T790M mutation in patients with acquired resistance to EGFR tyrosine kinase inhibitors. Lung Cancer 82: 294–298, https://doi.org/10.1016/j.lungcan.2013.08.023.Search in Google Scholar PubMed

Szymanowska, A., Jassem, E., Dziadziuszko, R., Skrzypski, M., Kobierska-Gulida, G., Holm, K., Borg, A., Rzyman, W., Limon, J., and Jassem, J. (2005). Analysis of prognostic value of TP53 gene mutations in non-small cell lung cancer. Pneumonol. Alergol. Pol. 73: 264–269.Search in Google Scholar

Tagliafierro, L. and Chiba-Falek, O. (2016). Up-regulation of SNCA gene expression: implications to synucleinopathies. Neurogenetics 17: 145–157, https://doi.org/10.1007/s10048-016-0478-0.Search in Google Scholar PubMed PubMed Central

Taira, T., Saito, Y., Niki, T., Iguchi-Ariga, S.M., Takahashi, K., and Ariga, H. (2004). DJ-1 has a role in antioxidative stress to prevent cell death. EMBO Rep. 5: 213–218, https://doi.org/10.1038/sj.embor.7400143.Search in Google Scholar

Takahashi-Niki, K., Ganaha, Y., Niki, T., Nakagawa, S., Kato-Ose, I., Iguchi-Ariga, S.M.M., and Ariga, H. (2016). DJ-1 activates SIRT1 through its direct binding to SIRT1. Biochem. Biophys. Res. Commun. 474: 131–136, https://doi.org/10.1016/j.bbrc.2016.04.084.Search in Google Scholar PubMed

Tanizaki, J., Okamoto, I., Okabe, T., Sakai, K., Tanaka, K., Hayashi, H., Kaneda, H., Takezawa, K., Kuwata, K., Yamaguchi, H., et al.. (2012). Activation of HER family signaling as a mechanism of acquired resistance to ALK inhibitors in EML4-ALK-positive non-small cell lung cancer. Clin. Cancer Res. 18: 6219–6226, https://doi.org/10.1158/1078-0432.ccr-12-0392.Search in Google Scholar PubMed

Testa, U., Castelli, G., and Pelosi, E. (2018). Lung cancers: molecular characterization, clonal Heterogeneity and evolution, and cancer stem cells. Cancers 10: 248, https://doi.org/10.3390/cancers10080248.Search in Google Scholar PubMed PubMed Central

Tian, Y., Lv, J., Su, Z., Wu, T., Li, X., Hu, X., Zhang, J., and Wu, L. (2021). LRRK2 plays essential roles in maintaining lung homeostasis and preventing the development of pulmonary fibrosis. Proc. Natl. Acad. Sci. U. S. A. 118: e2106685118, https://doi.org/10.1073/pnas.2106685118.Search in Google Scholar PubMed PubMed Central

Tolosa, E., Vila, M., Klein, C., and Rascol, O. (2020). LRRK2 in Parkinson disease: challenges of clinical trials. Nat. Rev. Neurol. 16: 97–107, https://doi.org/10.1038/s41582-019-0301-2.Search in Google Scholar PubMed

Tong, Y., Yamaguchi, H., Giaime, E., Boyle, S., Kopan, R., Kelleher, R.J.3rd, and Shen, J. (2010). Loss of leucine-rich repeat kinase 2 causes impairment of protein degradation pathways, accumulation of alpha-synuclein, and apoptotic cell death in aged mice. Proc. Natl. Acad. Sci. U. S. A. 107: 9879–9884, https://doi.org/10.1073/pnas.1004676107.Search in Google Scholar PubMed PubMed Central

Travis, W.D., Brambilla, E., Noguchi, M., Nicholson, A.G., Geisinger, K.R., Yatabe, Y., Beer, D.G., Powell, C.A., Riely, G.J., Van Schil, P.E., et al.. (2011). International association for the study of lung cancer/american thoracic society/european respiratory society international multidisciplinary classification of lung adenocarcinoma. J. Thorac. Oncol. 6: 244–285, https://doi.org/10.1097/JTO.0b013e318206a221.Search in Google Scholar PubMed PubMed Central

Tsujimoto, Y. and Shimizu, S. (2000). Bcl-2 family: life-or-death switch. FEBS Lett. 466: 6–10, https://doi.org/10.1016/s0014-5793(99)01761-5.Search in Google Scholar PubMed

Tysnes, O.-B. and Storstein, A. (2017). Epidemiology of Parkinson’s disease. J. Neural. Transm. 124: 901–905, https://doi.org/10.1007/s00702-017-1686-y.Search in Google Scholar PubMed

Vara-Perez, M., Felipe-Abrio, B., and Agostinis, P. (2019). Mitophagy in cancer: a tale of adaptation. Cells 8: 493, https://doi.org/10.3390/cells8050493.Search in Google Scholar PubMed PubMed Central

Vasseur, S., Afzal, S., Tardivel-Lacombe, J., Park, D.S., Iovanna, J.L., and Mak, T.W. (2009). DJ-1/PARK7 is an important mediator of hypoxia-induced cellular responses. Proc. Natl. Acad. Sci. U. S. A. 106: 1111–1116, https://doi.org/10.1073/pnas.0812745106.Search in Google Scholar PubMed PubMed Central

Vasseur, S., Afzal, S., Tomasini, R., Guillaumond, F., Tardivel-Lacombe, J., Mak, T.W., and Iovanna, J.L. (2012). Consequences of DJ-1 upregulation following p53 loss and cell transformation. Oncogene 31: 664–670, https://doi.org/10.1038/onc.2011.268.Search in Google Scholar PubMed

Veeriah, S., Taylor, B.S., Meng, S., Fang, F., Yilmaz, E., Vivanco, I., Janakiraman, M., Schultz, N., Hanrahan, A.J., Pao, W., et al.. (2010). Somatic mutations of the Parkinson’s disease-associated gene PARK2 in glioblastoma and other human malignancies. Nat. Genet. 42: 77–82, https://doi.org/10.1038/ng.491.Search in Google Scholar PubMed PubMed Central

Volpicelli-Daley, L.A., Abdelmotilib, H., Liu, Z., Stoyka, L.E., Daher, J.P.L., Milnerwood, A.J., Unni, V.K., Hirst, W.D., Yue, Z., Zhao, H.T., et al.. (2016). G2019S-LRRK2 expression augments α-synuclein sequestration into inclusions in neurons. J. Neurosci. 36: 7415–7427, https://doi.org/10.1523/jneurosci.3642-15.2016.Search in Google Scholar

Wallings, R., Manzoni, C., and Bandopadhyay, R. (2015). Cellular processes associated with LRRK2 function and dysfunction. FEBS J. 282: 2806–2826, https://doi.org/10.1111/febs.13305.Search in Google Scholar PubMed PubMed Central

Wang, P., Li, X., Li, X., Yang, W., and Yu, S. (2016). Blood plasma of patients with Parkinson’s disease increases alpha-synuclein aggregation and neurotoxicity. Parkinsons Dis. 2016: 7596482.10.1155/2016/7596482Search in Google Scholar PubMed PubMed Central

Wang, Q., Chen, Y., Feng, H., Zhang, B., and Wang, H. (2018). Prognostic and predictive value of HURP in nonsmall cell lung cancer. Oncol. Rep. 39: 1682–1692, https://doi.org/10.3892/or.2018.6280.Search in Google Scholar PubMed PubMed Central

Wang, Y., Liu, H.-H., Cao, Y.-T., Zhang, L.-L., Huang, F., and Yi, C. (2020). The role of mitochondrial dynamics and mitophagy in carcinogenesis, metastasis and therapy. Front. Cell Dev. Biol. 8: 413, https://doi.org/10.3389/fcell.2020.00413.Search in Google Scholar PubMed PubMed Central

Wang, Z., Liu, P., Inuzuka, H., and Wei, W. (2014). Roles of F-box proteins in cancer. Nat. Rev. Cancer 14: 233–247, https://doi.org/10.1038/nrc3700.Search in Google Scholar PubMed PubMed Central

Wei, Y., Chiang, W.C., Sumpter, R.Jr., Mishra, P., and Levine, B. (2017). Prohibitin 2 is an inner mitochondrial membrane mitophagy receptor. Cell 168: 224–238. e10, https://doi.org/10.1016/j.cell.2016.11.042.Search in Google Scholar PubMed PubMed Central

West, A.B., Dawson, V.L., and Dawson, T.M. (2005). To die or grow: Parkinson’s disease and cancer. Trends Neurosci. 28: 348–352, https://doi.org/10.1016/j.tins.2005.05.002.Search in Google Scholar PubMed

Wilkinson, K.D., Lee, K.M., Deshpande, S., Duerksen-Hughes, P., Boss, J.M., and Pohl, J. (1989). The neuron-specific protein PGP 9.5 is a ubiquitin carboxyl-terminal hydrolase. Science 246: 670–673, https://doi.org/10.1126/science.2530630.Search in Google Scholar PubMed

Xiong, D., Wang, Y., Kupert, E., Simpson, C., Pinney, S.M., Gaba, C.R., Mandal, D., Schwartz, A.G., Yang, P., De Andrade, M., et al.. (2015). A recurrent mutation in PARK2 is associated with familial lung cancer. Am. J. Hum. Genet. 96: 301–308, https://doi.org/10.1016/j.ajhg.2014.12.016.Search in Google Scholar PubMed PubMed Central

Xu, L., Lin, D.C., Yin, D., and Koeffler, H.P. (2014). An emerging role of PARK2 in cancer. J. Mol. Med. 92: 31–42, https://doi.org/10.1007/s00109-013-1107-0.Search in Google Scholar PubMed

Yan, Y., Xu, Z., Hu, X., Qian, L., Li, Z., Zhou, Y., Dai, S., Zeng, S., and Gong, Z. (2018). SNCA is a functionally low-expressed gene in lung adenocarcinoma. Genes 9: 16, https://doi.org/10.3390/genes9010016.Search in Google Scholar PubMed PubMed Central

Yang, C., Wang, H., Zhang, B., Chen, Y., Zhang, Y., Sun, X., Xiao, G., Nan, K., Ren, H., and Qin, S. (2016). LCL161 increases paclitaxel-induced apoptosis by degrading cIAP1 and cIAP2 in NSCLC. J. Exp. Clin. Cancer Res. 35: 158, https://doi.org/10.1186/s13046-016-0435-7.Search in Google Scholar PubMed PubMed Central

Yang, Y., Gehrke, S., Haque, M.E., Imai, Y., Kosek, J., Yang, L., Beal, M.F., Nishimura, I., Wakamatsu, K., Ito, S., et al.. (2005). Inactivation of Drosophila DJ-1 leads to impairments of oxidative stress response and phosphatidylinositol 3-kinase/Akt signaling. Proc. Natl. Acad. Sci. U. S. A. 102: 13670–13675, https://doi.org/10.1073/pnas.0504610102.Search in Google Scholar PubMed PubMed Central

Yao, J., Reyimu, A., Sun, A., Duoji, Z., Zhou, W., Liang, S., Hu, S., Wang, X., Dai, J., and Xu, X. (2022). UCHL1 acts as a potential oncogene and affects sensitivity of common anti-tumor drugs in lung adenocarcinoma. World J. Surg. Oncol. 20: 153, https://doi.org/10.1186/s12957-022-02620-3.Search in Google Scholar PubMed PubMed Central

Yilmaz, M., Wang, F., Loghavi, S., Bueso-Ramos, C., Gumbs, C., Little, L., Song, X., Zhang, J., Kadia, T., Borthakur, G., et al.. (2019). Late relapse in acute myeloid leukemia (AML): clonal evolution or therapy-related leukemia? Blood Cancer J. 9: 7, https://doi.org/10.1038/s41408-019-0170-3.Search in Google Scholar PubMed PubMed Central

Yoon, J.-H., Mo, J.-S., Kim, M.-Y., Ann, E.-J., Ahn, J.-S., Jo, E.-H., Lee, H.-J., Lee, Y.C., Seol, W., Yarmoluk, S.M., et al.. (2017). LRRK2 functions as a scaffolding kinase of ASK1-mediated neuronal cell death. Biochim. Biophys. Acta Mol. Cell Res. 1864: 2356–2368, https://doi.org/10.1016/j.bbamcr.2017.09.001.Search in Google Scholar PubMed

Zappa, C. and Mousa, S.A. (2016). Non-small cell lung cancer: current treatment and future advances. Transl. Lung Cancer Res. 5: 288–300, https://doi.org/10.21037/tlcr.2016.06.07.Search in Google Scholar PubMed PubMed Central

Zeng, H.Z., Qu, Y.Q., Zhang, W.J., Xiu, B., Deng, A.M., and Liang, A.B. (2011). Proteomic analysis identified DJ-1 as a cisplatin resistant marker in non-small cell lung cancer. Int. J. Mol. Sci. 12: 3489–3499, https://doi.org/10.3390/ijms12063489.Search in Google Scholar PubMed PubMed Central

Zhang, C.-W., Hang, L., Yao, T.-P., and Lim, K.-L. (2016). Parkin regulation and neurodegenerative disorders. Front. Aging Neurosci. 7, https://doi.org/10.3389/fnagi.2015.00248.Search in Google Scholar PubMed PubMed Central

Zhang, H., Yin, C., Liu, X., Bai, X., Wang, L., Xu, H., Ju, J., and Zhang, L. (2020a). Prohibitin 2/PHB2 in parkin-mediated mitophagy: a potential therapeutic target for non-small cell lung carcinoma. Med. Sci. Monit. 26: e923227, https://doi.org/10.12659/msm.923227.Search in Google Scholar

Zhang, L., Wang, J., Wang, J., Yang, B., He, Q., and Weng, Q. (2020b). Role of DJ-1 in immune and inflammatory diseases. Front. Immunol. 11: 994, https://doi.org/10.3389/fimmu.2020.00994.Search in Google Scholar PubMed PubMed Central

Zhang, R., Gu, J., Chen, J., Ni, J., Hung, J., Wang, Z., Zhang, X., Feng, J., and Ji, L. (2017). High expression of PINK1 promotes proliferation and chemoresistance of NSCLC. Oncol. Rep. 37: 2137–2146, https://doi.org/10.3892/or.2017.5486.Search in Google Scholar PubMed

Zhang, Y., Han, C.Y., Duan, F.G., Fan, X.-X., Yao, X.-J., Parks, R.J., Tang, Y.-J., Wang, M.-F., Liu, L., Tsang, B.K., et al.. (2019). p53 sensitizes chemoresistant non-small cell lung cancer via elevation of reactive oxygen species and suppression of EGFR/PI3K/AKT signaling. Cancer Cell Int. 19: 188, https://doi.org/10.1186/s12935-019-0910-2.Search in Google Scholar PubMed PubMed Central

Zhao, C., He, R., Shen, M., Zhu, F., Wang, M., Liu, Y., Chen, H., Li, X., and Qin, R. (2019). PINK1/Parkin-Mediated mitophagy regulation by reactive oxygen species alleviates rocaglamide A-induced apoptosis in pancreatic cancer cells. Front. Pharmacol. 10, https://doi.org/10.3389/fphar.2019.00968.Search in Google Scholar PubMed PubMed Central

Zheng, H., Zhou, C., Lu, X., Liu, Q., Liu, M., Chen, G., Chen, W., Wang, S., and Qiu, Y. (2018). DJ-1 promotes survival of human colon cancer cells under hypoxia by modulating HIF-1alpha expression through the PI3K-AKT pathway. Cancer Manag. Res. 10: 4615–4629, https://doi.org/10.2147/cmar.s172008.Search in Google Scholar

Zhong, H., De Marzo, A.M., Laughner, E., Lim, M., Hilton, D.A., Zagzag, D., Buechler, P., Isaacs, W.B., Semenza, G.L., and Simons, J.W. (1999). Overexpression of hypoxia-inducible factor 1alpha in common human cancers and their metastases. Cancer Res. 59: 5830–5835.Search in Google Scholar

Zhou, Z.D., Sathiyamoorthy, S., Angeles, D.C., and Tan, E.K. (2016). Linking F-box protein 7 and parkin to neuronal degeneration in Parkinson’s disease (PD). Mol. Brain 9: 41, https://doi.org/10.1186/s13041-016-0218-2.Search in Google Scholar PubMed PubMed Central

Zhou, Z.D., Xie, S.P., Sathiyamoorthy, S., Saw, W.T., Sing, T.Y., Ng, S.H., Chua, H.P.H., Tang, A.M.Y., Shaffra, F., Li, Z., et al.. (2015). F-box protein 7 mutations promote protein aggregation in mitochondria and inhibit mitophagy. Hum. Mol. Genet. 24: 6314–6330, https://doi.org/10.1093/hmg/ddv340.Search in Google Scholar PubMed

Zimprich, A., Biskup, S., Leitner, P., Lichtner, P., Farrer, M., Lincoln, S., Kachergus, J., Hulihan, M., Uitti, R.J., Calne, D.B., et al.. (2004). Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron 44: 601–607, https://doi.org/10.1016/j.neuron.2004.11.005.Search in Google Scholar PubMed

Received: 2022-07-12
Accepted: 2022-12-13
Published Online: 2023-01-13
Published in Print: 2023-05-25

© 2022 the author(s), published by De Gruyter, Berlin/Boston

This work is licensed under the Creative Commons Attribution 4.0 International License.

Downloaded on 7.1.2026 from https://www.degruyterbrill.com/document/doi/10.1515/hsz-2022-0228/html
Scroll to top button