Disruption of the circadian clock circuitry plays a crucial role in cancer onset and progression [
69,
70]. Derangement of the biological clock and single polymorphisms of some circadian genes have been linked to cancer susceptibility [
71]. NPAS2 is regarded as a promising predictor of clinical outcome in various malignancies and was reported to behave as tumor suppressor in colorectal carcinogenesis [
72]. DNA CpG methylation profile and chromatin modification patterns represent fundamental constituents of the cell epigenome and chromatin regulators are major controllers of gene transcription in normal cells, directing histone modifications and chromatin remodeling.
The biological clock drives cycles in spatial and temporal chromosomal organization controlling rhythmicity of transcriptional activation and repression [
73]
. During carcinogenesis genetic mutations and/or epigenetic alterations disrupt the functions of chromatin regulators initiating wide-ranging derangement of gene expression [
74]. In this regard, NPAS2 was identified as component of a four chromatin regulators hub (CBX7, HMGA2, NPAS2 and PRC1) allowing risk stratification and outcome prediction in lung adenocarcinoma patients and providing a gene signature enriching PI3K/Akt/mTOR pathway and p53 signaling and associated to the infiltration percentages of macrophage M0, macrophage M2, resting NK cells, memory B cells, dendritic cells in tumor microenvironment [
75]. Accordingly,
NPAS2 was comprised in a five-gene signature (
DKK1,
CCL20,
NPAS2,
GNPNAT1 and
MELTF) predicting prognosis (decreased overall survival rates and shorter progression-free survival) and immunotherapy response of lung adenocarcinoma patients [
76]. Likewise, higher expression of the NPAS2 gene in tumor tissue was closely related with immune infiltration and overall survival of glioma patients [
77] as well as to immune infiltration and poor prognosis in hepatocellular carcinoma patients [
78,
79]. In the setting of hepatocellular carcinogenesis, upregulation of NPAS2 expression, chiefly attributable to the downregulation of miR-199b-5p, was found capable to reprogram glucose metabolism through transcriptional activation of HIF-1α with upregulation of the glycolytic genes
GLUT1,
HK2,
GPI,
ALDOA,
ENO2,
PKM2 and
MCT4 and downregulation of peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α) with subsequent reduced mitochondrial biogenesis [
80]. Interestingly, NPAS2 was found upregulated in hepatic stellate cells (HSCs) after fibrogenic injury, with ensuing HSCs activation, capable to play a role in liver fibrogenesis though direct transcriptional activation in HSCs of hairy and enhancer of split 1 (Hes1), a critical transcription factor involved in Notch signaling [
81]. Additionally, in anaplastic thyroid carcinoma NPAS2 was found to be significantly up-regulated in tumor tissue and
in vitro experiments showed that NPAS2 silencing in anaplastic thyroid carcinoma cell lines successfully obstructed cell proliferation, migration and invasion [
82]. In the context of hormone-sensitive cancers, NPAS2 gene down-regulation was found associated with poorly differentiated tumors in comparison with well and moderately differentiated breast cancer [
83] and rs2305160 polymorphism of NPAS2 gene (Ala394Thr) was found strongly associated with breast cancer risk and proposed as candidate breast cancer susceptibility locus [
84,
85,
86]. Remarkably, Ala394Thr polymorphism in the NPAS2 gene was identified as modifier for the risk of non-Hodgkin's lymphoma, with robust association of the variant Thr genotypes (Ala/Thr and Thr/Thr) with reduced risk of lymphomagenesis [
87]. Regarding other hormone-dependent tumors, NPAS2 dysregulation was found associated with poor prognosis and clinic-pathological characteristics of patients suffering from uterine corpus endometrial carcinoma (UCEC). NPAS2 upregulation in UCEC tissue samples compared to matched non-tumorous tissue was associated with higher clinical stage and tumor grade, estrogen receptor status, myometrial invasion, leading to decreased overall, disease free and relapse free survival in UCEC patients. Besides, NPAS2 showed significant negative correlations with miR-17-5p and miR-93-5p and positive correlations with miR106a-5p and miR-381-3p and NPAS2 knockdown or overexpression
in vitro in UCEC cell lines impacted cell proliferation and apoptosis [
88]. Among malignant genito-urinary tract neoplasms, NPAS2 expression in prostate neoplasia suggested tumor suppressor function and showed significant correlation with the immune components of the tumor microenvironment and was identified as part of a gene signature capable to predict probability of disease progression in prostate cancer patients [
89]. In tumor tissue of prostate cancer patients NPAS2 expression was found upregulated respect to matched non-tumorous tissue. NPAS2 knockdown in prostate cancer cell lines hindered cell proliferation and upholded cell apoptosis
in vitro and restrained tumor growth
in vivo in xenotransplanted nude mice. At the molecular level, NPAS2 supported glycolysis and restrained oxidative phosphorylation in prostate cancer cell lines through amplification of hypoxia-inducible factor-1A (HIF-1A)-dependent signaling. Accordingly, NPAS2 knockdown decreased glucose uptake and lactate production, while increasing intracellular pH and oxygen consumption rate [
90]. An association between NPAS2 expression and prostate cancer was also corroborated in the EPICAP study, a population-based case-control study with genotyped data that investigated the relationship between circadian genes polymorphisms and prostate cancer risk [
91]. The non-synonymous mutations and genetic variants rs1369481, rs895521, and rs17024926 in NPAS2 gene were found significantly associated with susceptibility to prostate cancer, considering overall risk and risk of aggressive disease [
92]. Moreover, the NPAS2 gene single nucleotide polymorphism rs6542993 A>T was found significantly associated with higher risk of disease progression in both localized and advanced prostate cancer cases, with decreased NPAS2 expression levels found in carriers of the T allele of rs6542993 compared with those carrying the A allele and associated with more aggressive prostate cancer and poor progression-free survival [
93]. On the other hand, NPAS2 genetic variant rs895520 was found associated with a statistically significant higher predisposition to sarcoma and leiomyosarcoma [
94]. Distinct expression of luminal and basal gene features muscle-invasive bladder cancers, practical biomarkers to predict disease progression and overall survival. NPAS2 was found to be clinically relevant and involved in the regulation of subtype-specific genes influencing cancer cell proliferation and migration in luminal bladder cancers [
95]. As regards myeloproliferative neoplasms, NPAS2 was found upregulated in acute myeloid leukemia cells and experiments performed both in vivo and in vitro highlighted its fundamental role in supporting cell survival and proliferation as well as in restraining apoptosis at various stages of myeloid differentiation. NPAS2 knockdown obstructed CDC25A expression leading to G1/S cell cycle arrest, augmented caspase-3 cleavage and promoted cell death through Bcl2/Bax production modification [
96]. A similar survival-promoting role was found for NPAS2 in HCC through transcriptional activation of the CDC25A phosphatase and consequent dephosphorylation of CDK2/4/6 and Bcl-2, which prompted cell proliferation and obstructed cell apoptosis, correspondingly [
97]. MicroRNAs (miRNAs) are small, single-stranded, non-coding RNA molecules containing 21 to 23 nucleotides that post-transcriptionally regulate expression of coding genes in a sequence-specific manner. MicroRNAs modulate oscillating mRNA levels and are essential for generating a time delay critical for the molecular clockwork [
98]. MicroRNAs impact cancer onset and progression as well as response to therapy modifying gene expression. RNA-seq and miRNA-omics analysis performed in radio-resistant nasopharyngeal cancer (NPC) cells pinpointed NPAS2-miR-20a-5p axis as crucially involved in NPC radio-resistance and this role was corroborated by experiments with down- or up-regulation of their levels in NPC cell lines [
99]. Sequencing of putative promoter and 5' untranslated region of the NPAS2 promoter in patients suffering from melanoma identified several variants. Of particular interest was a microsatellite comprising a GGC repeat with different alleles ranging from 7 to 13 repeats located in the 5' untranslated exon. Homozygosity of an allele with nine repeats (9/9) was more prevalent in melanoma patients respect to control subjects, suggesting a role for NPAS2 variants in melanoma susceptibility [
100].