Next Article in Journal
Integrins as Key Mediators of Metastasis
Previous Article in Journal
Co-Location of QTL for Vigor and Resistance to Three Diseases in Juglans microcarpa × J. regia Rootstocks
Previous Article in Special Issue
A Senescent Cluster in Aged Human Hematopoietic Stem Cell Compartment as Target for Senotherapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Preeclampsia as a Study Model for Aging: The Klotho Gene Paradigm

by
Monia Cecati
1,
Stefania Fumarola
2,
Salvatore Vaiasicca
2,
Laura Cianfruglia
2,
Arianna Vignini
3,*,
Stefano Raffaele Giannubilo
4,*,
Monica Emanuelli
3,† and
Andrea Ciavattini
4,†
1
Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy
2
Scientific Direction, IRCCS INRCA, 60124 Ancona, Italy
3
Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica Delle Marche, 60126 Ancona, Italy
4
Department of Clinical Sciences, Clinic of Obstetrics and Gynaecology, Università Politecnica Delle Marche, 60123 Ancona, Italy
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work as joint senior authors.
Int. J. Mol. Sci. 2025, 26(3), 902; https://doi.org/10.3390/ijms26030902
Submission received: 28 December 2024 / Revised: 18 January 2025 / Accepted: 20 January 2025 / Published: 22 January 2025

Abstract

:
Aging and pregnancy are often considered opposites in a woman’s biological timeline. Aging is defined by a gradual decline in the functional capabilities of an organism over its lifetime, while pregnancy is characterized by the presence of the transient placenta, which fosters the cellular fitness necessary to support fetal growth. However, in the context of preeclampsia, pregnancy and aging share common hallmarks, including clinical complications, altered cellular phenotypes, and heightened oxidative stress. Furthermore, women with pregnancies complicated by preeclampsia tend to experience age-related disorders earlier than those with healthy pregnancies. Klotho, a gene discovered fortuitously in 1997 by researchers studying aging mechanisms, is primarily expressed in the kidneys but also to a lesser extent in several other tissues, including the placenta. The Klotho protein is a membrane-bound protein that, upon cleavage by ADAM10/17, is released into the circulation as soluble Klotho (sKlotho) where it plays a role in modulating oxidative stress. This review focuses on the involvement of sKlotho in the development of preeclampsia and age-related disorders, as well as the expression of the recently discovered Mytho gene, which has been associated with skeletal muscle atrophy.

1. Introduction

Preeclampsia is a placenta-mediated, multisystem hypertensive disorder of pregnancy that affects 2–8% of pregnancies [1]. In its severe form, it affects about 1.4% of pregnancies in the Western world, is characterized by maternal organ damage and/or fetal growth restriction, and is a major cause of maternal and infant morbidity and mortality [2,3].
One year after pregnancy, women have higher cardiovascular risks, such as hypertension, obesity, and dyslipidemia, making them 2–7 times more likely to develop cardiovascular diseases (CVDs) early compared to those with a normotensive pregnancy [4,5]. Women with preeclampsia defined as “early” preeclampsia—that is, at onset before 34 weeks’ gestation—have an even higher risk of up to 9 times higher risk of death from cardiovascular disease [6]. Preeclampsia is characterized by increased oxidative stress, vascular damage, and endothelial and metabolic dysfunctions [7,8,9,10,11].
Oxidative stress can activate several transcription factors, which lead to the expression of genes involved in the activation of inflammatory pathways favoring the production of inflammatory cytokines and the establishment of an inflammatory environment, which is involved in the onset and progression of several diseases [12,13,14,15,16,17,18].
The increased oxidative stress, due to placental hypoxia, is also involved in the chronic inflammatory state characterizing preeclamptic pregnancies [19]. Although preeclampsia and cardiovascular disease share common risk factors such as hypertension and obesity [20], there is evidence that preeclampsia is accompanied by vascular biochemical dysfunction that lingers throughout a woman’s life and, in later life, manifests itself as a potentially life-threatening disease [21,22]. Many studies in recent years have hypothesized and demonstrated genetic variants that may influence the risk of developing preeclampsia [23]. The heritability of preeclampsia is estimated at about 55% [24,25]. Genome-wide association studies (GWASs) [26] have reported maternal and fetal heritability for Europe as 38.1% and 21.3%, respectively, while those for Central Asia have reported 54.4% and 42.5%, respectively. In addition to the maternal association, it is also necessary to consider the fetal/neonatal association to future cardiovascular disease. The incidence of cardiovascular morbidity is significantly higher among infants born to preeclamptic women [27]. The pathophysiological basis of preeclampsia in fact lies in the altered invasion of the trophoblast and remodeling of the spiral artery, resulting in the restriction of uteroplacental circulation [28,29]. The fetus of a preeclamptic woman is in a setting of multiple insults, including ischemia, hypoxia, placental insufficiency, imbalance of angiogenic factors, inflammation, and oxidative stress [30]. The fetuses of preeclamptic women exhibit microcirculation abnormalities, endothelial disfunction, and accelerated progression of atherosclerosis [31]. The mechanisms connecting in utero exposure to preeclampsia with an increased risk of future cardiovascular disease may involve a complex interaction between shared genetic factors, environmental influences, and developmental programming. CVDs are also closely linked to the aging process. Specifically, in elderly women (>60 years) the prevalence of CVDs is even higher than in men [32].
At first glance, pregnancy and aging may appear to be opposing processes: pregnancy is characterized by the presence of the transient placenta, which develops the cellular fitness required to support fetal growth, while aging is marked by a progressive decline in cellular function. However, when considering preeclampsia, both pregnancy and aging may share common cellular hallmarks, which will be explored further in this paper.

2. Methods

To identify relevant studies, we conducted a comprehensive literature search across several databases, including PubMed, Scopus, and Web of Science. The search aimed to locate significant English-language research articles and reviews published in peer-reviewed journals over the past two decades (2000–2024). However, we also included foundational studies on preeclampsia and the Klotho gene published before this period. Our focus was on both in vivo human and animal studies to ensure clinical relevance while excluding studies lacking significant outcomes related to preeclamptic pregnancies or age-related disorders.
The search terms were carefully selected to encompass a wide range of research on the relationship between Klotho expression and preeclampsia or aging. The following keywords and phrases, in various combinations, were used: ‘Klotho’, ‘Healthy pregnancies’, ‘Preeclamptic pregnancies’, ‘Age-related disorders’, ‘Epigenetic modifications’, ‘Klotho expression in pregnancies’, ‘Klotho dysregulation in preeclamptic pregnancies’, and ‘Klotho dysregulation in age-related disorders’. Boolean operators (AND, OR, NOT) were applied to refine and expand the search.
Inclusion criteria focused on studies exploring the effects of Klotho administration in cell cultures or animal models, as well as research on peripheral Klotho concentrations or tissue expression in both young and elderly humans. Exclusion criteria ruled out non-English studies, articles published before 1994, and those lacking relevant clinical or experimental data. This approach ensured a thorough and wide-reaching review, emphasizing studies that provided clinical insights and a mechanistic understanding of the relationship between Klotho, preeclampsia, and age-related disorders.

3. Preeclampsia, Aging and Cellular Phenotype

Aging is characterized by a gradual decline in the functional capabilities of an organism over the course of its life. During the aging process, multiple organs undergo progressive deterioration of their physiological functions, leading to the onset of various diseases [33].
To date, impaired angiogenesis [34] and inflammation [35] are the two mechanisms known to be the most responsible for the pathophysiology of preeclampsia. The poor trophoblast invasion of the spiral arteries [28,36,37] in the placenta bed compromises placentation [38] and vascularization [39], induces an inappropriate activation of the immune system [40], and results in an inevitable dysregulation of several metabolic pathways [41,42]. The inability of preeclamptic tissue to return to a physiological state initiates an accelerated aging process, driving the placenta toward premature senescence and impairing its ability to sustain fetal growth until the normal conclusion of pregnancy. Cellular senescence is one of the aging hallmarks. Senescent cells were discovered by Hayflick and Moorhead in 1961 [43] and were later defined as cells that exit the cell cycle and enter replicative senescence due to the loss of their proliferative potential [44]. Telomere shortening is a hallmark of cellular senescence. In humans, telomeres consist of tandem repeats of the DNA sequence (TTAGGG) at the ends of chromosomes. These telomeres play a crucial role in maintaining chromosome stability and protecting coding DNA from recombination, degradation, and replication damage, a function recognized for many years.
In adult human cells, the reduced activity of DNA telomerase leads to telomere shortening. With each cell division, telomeres become progressively shorter. To prevent telomere length from falling below a critical threshold, a permanent cell cycle arrest occurs. Shortened telomeres are associated with age-related diseases, including metabolic disorders (e.g., metabolic syndrome, liver cirrhosis) and cardiovascular conditions (e.g., atherosclerosis) [45,46].
Telomeres in preeclamptic trophoblast tissue are shorter compared to those in physiological trophoblast tissue [47,48]. In preeclampsia, excessive fetal hypoxia during the early stages of placentation induces DNA damage in the telomeric region. This dysfunctional telomere length, combined with reduced telomerase activity, may contribute to the accelerated placental aging observed in preeclamptic pregnancies [49].
Telomerase activity and the maintenance of telomere length are known to be linked to the immortality of cancer cells, germline cells, and embryonic stem cells [50]. The expression of genes related to undifferentiated embryonic stem cells is increasingly being used as a biomarker for tissue aging in a growing number of studies [51,52,53]. A reduced expression of stem cell markers is reported in trophoblast tissue from preeclamptic pregnancy [47] but also in aging tissue as synonyms of decline in tissue regenerative potential [54].
Although quiescent in the cell cycle, senescent cells undergo significant changes in their gene expression profile. Specifically, they are characterized by the secretion of cytokines, chemokines, and proteases with pro-inflammatory properties, a phenomenon known as the senescence-associated secretory phenotype (SASP) [55].
Strong evidence indicates that the senescence-associated secretory phenotype (SASP) is closely associated with critical clinical parameters and outcomes, with its impact intensifying with age. This is likely a result of the growing accumulation of senescent cells in one or more organs as biological age progresses [56]. Published studies also highlight dysregulated levels of pro-inflammatory cytokines associated with SASP in older individuals.
The literature reports age-related increments of IL-6 [57], IL-8 [58], monocyte chemoattractant protein 1 (MCP-1) [55], and plasminogen activator inhibitor 1 (PAI-1) [59] in blood concentrations from elderly women. Additionally, IL-6 levels in venous blood from older women (over 65 years of age) are positively correlated with high blood pressure and negatively correlated with estimated glomerular filtration rate (eGFR), a test used to assess kidney function and determine the stage of kidney disease [60]. Notably, serum IL-8 levels were significantly increased in patients affected by chronic liver disease, with respect to healthy subjects [61]. Duisenbek et al. reported that MCP-1 levels were higher in the blood of elderly patients affected by type 2 diabetes and cardiovascular disease than that of healthy subjects [62].
Stubblefield et al. demonstrated that PAI-1 was significantly increased in patients affected by type 2 diabetes and metabolic syndrome when compared with disease-negative controls [63]. Type 2 diabetes, cardiovascular diseases, and kidney and liver failure are clinical hallmarks of both aging and preeclampsia. In a study by Suvakov et al., patients with pregnancies complicated by preeclampsia were recruited. They demonstrated that women with preeclampsia had elevated levels of inflammatory mediators (e.g., IL-6, IL-8, MCP-1, and PAI-1) in both blood and adipose tissue compared to women with healthy pregnancies [64].
Aging also impacts mitochondrial function, diminishing the efficiency of oxidative phosphorylation. This decline is linked to altered ATP production and an increase in the generation of reactive oxygen species (ROS) [65]. Excessive ROS can lead to the oxidation of mitochondrial DNA, proteins, and lipids, impairing the cell’s ability to activate mitophagy and ultimately resulting in mitochondrial dysfunction [66,67].
Studies have shown that mitochondrial dysfunction in elderly women is associated with cardiovascular disease [68], increased risk of cardiovascular disease [69], and metabolic syndrome [70].
The content of extracellular vesicles released into the circulation in mitochondrial DNA (mtDNA) was higher in frail elderly women compared to healthy subjects [71].
Since the late 1980s, mitochondrial disorders have been identified as potential contributors to preeclampsia [72,73]. Prolonged hypoxia or recurrent cycles of ischemia/reoxygenation during preeclampsia can further exacerbate mitochondrial dysfunction [74]. Serum levels of mtDNA have been shown to be elevated in pregnancies complicated by preeclampsia compared to healthy pregnancies [75].
mtDNA is not the only circulating biomarker associated with aging and preeclampsia. Circulating microRNAs (miRNAs) also play a significant role in the pathogenesis, diagnosis, and prediction of both preeclampsia and aging. Given their potential therapeutic implications for treating preeclampsia and age-related diseases, the following section will focus on miRNAs, as well as on two other key epigenetic modifications: DNA methylation and histone modification.

4. Preeclampsia, Aging, and Epigenetics

The term “epigenetics” was first introduced in the early 1940s, and its meaning has evolved significantly since Conrad Waddington’s definition in 1968 [76]. Today, epigenetics is generally understood as “the study of changes in gene function that are mitotically and/or meiotically heritable and that do not involve a change in the DNA sequence” [77]. The three most studied epigenetic modifications are DNA methylation, microRNAs (miRNAs), and histone modification. Abnormal epigenetic modifications to DNA have been observed in cells from various sources, not only in elderly individuals but also in pregnant women with preeclampsia. In this section, we aim to provide a concise overview of the existing literature, focusing on epigenetic modifications in peripheral blood cells to highlight the similarities between aging and preeclampsia. DNA methylation is a heritable epigenetic modification that involves the covalent transfer of a methyl group to the C-5 position of the cytosine ring in DNA [78].
This reaction is catalyzed by DNA methyltransferases (DNMTs), which use S-adenosylmethionine (SAM) as the methyl donor. SAM, however, also serves as a co-substrate for many other methyltransferases [79]. DNA methylation is mediated by a family of DNMTs: DNMT1, DNMT2, and the maintenance DNMTs, along with DNMT3A, DNMT3B, and DNMT3L, which are referred to as de novo DNMTs [80,81].
In somatic cells, more than 98% of DNA methylation occurs in CpG-rich regions of the genome [80]. DNMT1 and DNMT2 primarily methylate hemimethylated DNA in vitro and are active during the S phase of replication. In contrast, DNMT3A, DNMT3B, and DNMT3L prefer unmethylated CpG dinucleotides and are involved in de novo methylation during development [80,81].
In 2014, White et al. demonstrated that maternal leukocyte DNA methylation was altered in pregnancies with preeclampsia compared to healthy pregnancies. Specifically, differential methylation of certain neuronal genes, including GRIN2b, GABRA1, PCDHB7, and BEX1, was associated with an increased risk of preeclampsia [82]. In 2016, Rahat et al. studied the methylation level in the STAT5A promoter region by recovering cell-free fetal DNA from maternal plasma in both preeclamptic and healthy pregnancies. The study showed that hypermethylation of the STAT5A promoter region was positively correlated with the onset of preeclampsia [83]. Additionally, Zakeri et al. used the methylation-specific PCR method to identify DNA hypermethylation in the promoter region of the Nrf2 gene in the peripheral blood of preeclamptic individuals [84]. Hypermethylation in the promoter regions of both p21 and TP53 genes was also linked to preeclampsia [85].
Weiping et al. demonstrated that preeclampsia is associated with reduced methylation levels in the promoter region of the GNA12 gene. Furthermore, the methylation level of the GNA12 promoter was found to be similar in both the placenta and peripheral blood of pregnant women [86]. Lu et al. also identified an association between preeclampsia and hypomethylation in the promoter region of the CTGF gene, observed in both placentas and peripheral blood from a Chinese population [87]. Maternal vascular dysfunction, a hallmark of preeclampsia, has been linked to DNA methylation changes. Mousa et al. and Walsh et al. demonstrated that the methylation levels in the promoter regions of TBXAS1 [88], MMP-1 [89], and IL-17 cytokines [90] were reduced in the systemic blood vessels of preeclamptic women. Several of the genes discussed above also exhibit similar methylation patterns in age-related diseases. Hypermethylation in the promoter of the p21 gene has been strongly correlated with acute lymphoblastic leukemia and poor prognosis in elderly women [91]. In patients with type 2 diabetes mellitus, with or without nephropathy, methylation levels in the promoter region of the CTGF gene were significantly lower compared to the control group [92]. Additionally, a significantly decreased methylation level in the IL-17 promoter was observed in patients with age-related macular degeneration [93].
Aberrant DNA methylation (hypermethylation or hypomethylation) at promoter CpG islands leads to transcriptional silencing or the overexpression of genes and miRNAs. This disruption can cause the improper execution of key cellular pathways, much like genetic changes (e.g., mutations or deletions) [94].
miRNAs are single-stranded, non-coding RNAs, typically 20–24 nucleotides in length, derived from the primary miRNA transcript (pri-miRNA) [95]. The pri-miRNA is enzymatically processed, and the resulting mature miRNA is incorporated into the RNA-induced silencing complex (RISC) with the Argonaute2 protein, which induces mRNA target silencing [96,97]. Once released from the cell, often within extracellular vesicles such as exosomes and microvesicles [98,99], miRNAs exert their suppressive function by targeting mRNA, reaching recipient cells through peripheral circulation [100,101]. Due to their abundant presence in all body fluids, miRNAs serve as valuable biomarkers for a variety of diseases, including preeclampsia and aging [102,103]. Wang et al. isolated circulating exosomes from the peripheral blood of preeclamptic women and found an upregulation of miR-15a-5p compared to healthy pregnant women [104]. Circulating miR-132 [105] and miR-151a-3p [106] were significantly higher in preeclamptic women than in controls. A decrease in miR-93-5p and miR-126-3p levels in peripheral circulation, when associated with the overexpression of miR-125, increased the risk of preeclampsia. miR-125 inhibits cytotrophoblast invasion and impairs endothelial cell function [104]. The apoptosis-related miR-21 was overexpressed in the maternal plasma of women with preeclampsia and has been used as a biomarker to distinguish between mild and severe preeclampsia [107].
miR-15a-5p [108] and miR-151a-3p [109] were also found to be upregulated in the peripheral blood of elderly women with cognitive decline. Elderly women with mild cognitive impairment showed an increased expression of miR-132 compared to age-matched controls [110]. Kocijan et al. investigated the circulating miRNA profile in the peripheral blood of patients with idiopathic and postmenopausal osteoporosis and fragility fractures, suggesting miR-93-5p as a potential biomarker for bone-related pathologies [111]. Circulating miR-126-3p levels were lower in the peripheral blood of 246 hospitalized geriatric patients with cardiovascular diseases and multimorbidity and were positively associated with a higher risk of short- and medium-term mortality [112].
Olivieri et al. conducted a miRNA plasma profiling study with 11 healthy individuals aged 20, 80, and 100 years. They validated the selected miRNAs in a cohort of 111 healthy adults aged 20–105 years and 30 centenarians. Additionally, 34 patients with CVDs and 15 healthy centenarian offspring (CO) were included in the study. The results showed that miR-21 expression was higher in CVD patients and lower in CO compared to age-matched controls [113].
Histones, the protein components of chromatin, undergo posttranslational modifications that influence chromatin structure [114]. Histone acetylation and histone methylation are the two most widely studied modifications in the context of preeclampsia pathology. Histone acetylation is catalyzed by histone acetyltransferases and leads to gene transcriptional activation [115] by making the DNA–protein complex more accessible to transcription factors and RNA polymerase II [116].
Unlike acetylation, the methylation of lysine residues can have varying effects on gene expression, depending on the specific lysine residue that undergoes methylation. Among the identified methylated lysine residues, several have been extensively studied. For example, methylation at lysine 4 (K4) of histone 3 (H3) promotes gene expression, while methylation at K9 of H3 is linked to transcriptional repression [117].
Whole genome analysis of cytotrophoblasts from severe preeclampsia revealed a significant increase in H3K27 acetylation. Interestingly, genes that are typically downregulated during a physiological pregnancy at term were found to be upregulated in this syndrome [118].
When analyzing the expression of histone proteins H3K4me3 and H3K9ac in both preeclamptic and control placentas through immunohistochemistry, Meister et al. observed a significant global reduction in trimethylated histone H3K4me3 and acetylated H3K9ac in preeclamptic tissue. These findings suggested a loss of accessibility for genes to be transcribed [118,119]. Sheng et al. specifically evaluated histone H3K9 methylation protein expression in HUVECs from normal and preeclamptic pregnancies. Their data showed a strong correlation between di- and tri-H3K9 methylation and reduced antioxidant CuZn-SOD expression in fetal endothelial cells from preeclampsia, reinforcing the critical role of oxidative stress in the development of this syndrome [120].
Histone methylation is closely related to oxidative stress. Whongsiri et al. demonstrated that in bladder cancer tissues, an increase in 8-OHdG was associated with elevated tri-methylated H3K9, supporting the hypothesis that oxidative stress also influences genome reprogramming [121]. Aging has also been linked to defective muscle regeneration due to a reduction in H3K9 di/tri-methylation [122]. In esophageal squamous cell carcinoma, Chen et al. observed the global hypoacetylation of H3 and H4, along with hypermethylation of H3K4 and H3K27. Furthermore, these modifications were correlated with the severity of the disease [123]. Oxidative stress plays a pivotal role in regulating the system of epigenetic modifications and contributes to the pathophysiology of age-related disorders and preeclampsia, as discussed by the authors [124,125].

5. Oxidative Stress in Preeclampsia and Aging

Oxidative stress results from an imbalance between reactive oxygen species (ROS) and antioxidants. ROS are primarily oxygen-based molecules, including the superoxide anion radical (O2), hydrogen peroxide (H2O2), and hydroxyl radicals [126]. These reactive species can structurally damage cellular compounds such as DNA, lipids, carbohydrates, and proteins, necessitating a balance with antioxidant defenses [126,127,128,129]. Under physiological conditions, ROS function as signaling molecules [130,131] playing a role in mediating maternal immune tolerance to the semi-allogenic fetus [132], similar to the role of adenosine [133].
Oxidative stress is implicated in the onset and progression of various cancerous and non-cancerous diseases [134,135,136,137,138].
It also regulates key signaling networks, including forkhead transcription factors of the O class (FoxO) [139]. During a healthy pregnancy, the development of fetal organs and tissues leads to increased metabolic activity and oxygen consumption. Fatty acids are utilized to produce energy for sustaining maternal retroplacental tissues. This process inevitably generates ROS, particularly H2O2. However, in a physiological pregnancy, oxidative stress triggers antioxidant mechanisms that modulate enzyme activity and interact with non-enzymatic free radical deactivators, helping to eliminate excess ROS and maintain cellular homeostasis [140]. Pregnancy is a state where the balance between ROS and antioxidant defenses can be easily disrupted. Under pathological conditions, elevated ROS levels can stimulate pathological redox signaling, leading to cellular damage and contributing to various disease states [141,142]. Studies focused on pregnancy have shown that oxidative stress is associated with complications during pregnancy and may impact fetal development [143]. Preeclampsia is a pregnancy-related pathology linked to oxidative stress. Although the exact etiology of preeclampsia remains unclear, the underlying pathological event appears to be endothelial injury, which results from elevated placental ROS levels or reduced antioxidant activity [144,145]. This injury leads to hypoxia, which triggers the conversion of xanthine dehydrogenase to xanthine oxidase, further increasing ROS production in the placenta [146,147]. Preeclamptic placentas are also characterized by mitochondrial abnormalities and disruptions in the molecular pathways regulating mitochondrial function [148].
Aging is closely linked to oxidative stress [149], as the increased production of ROS during aging is primarily attributed to mitochondrial dysfunction. This creates a vicious cycle, where oxidative stress exacerbates mitochondrial dysfunction [150]. Oxidative stress plays a significant role in the age-related decline of organ function, including kidney function. Chronic kidney disease (CKD) becomes more prevalent with age, affecting 27.9% of individuals over 70, compared to only 8.1% in the general population [151]. Kidney tubular cells are particularly sensitive to oxidative stress, adopting a senescent phenotype [152]. Autophagy is a key mechanism by which proximal tubular cells maintain homeostasis and protect the kidneys from injury. Yamamoto et al. observed that inadequate autophagic flux in response to metabolic stress increases the risk of age-related kidney diseases [153]. Oxidative stress and imbalanced ROS levels are also implicated in the development of Alzheimer’s disease in aging brains. Martin-Maestro et al. studied mitophagy in hippocampal samples from Alzheimer’s patients, finding that alterations in the mitophagy process were strongly associated with increased ROS production and clinical manifestations of the disease [154]. In human lung epithelial A549 cells, exposure to a high phosphate medium resulted in elevated ROS levels, and the inhibition of antioxidant pathways led to apoptosis of the A549 cells [155].

6. The Klotho Gene

In 1997, while exploring the human genome, a research group led by Kuro-o discovered a gene by chance. As their focus was on aging mechanisms, Kuro-o decided to name the gene Klotho, after the Greek goddess Clotho, who spins the thread of life [156]. The Klotho gene in humans is located on chromosome 13q12 and spans over 50 kb.
Subsequently, the name α-Klotho was introduced to refer to the “original” Klotho gene in humans, distinguishing it from its homolog in mice, which was named β-Klotho [157]. In humans, β-Klotho shares 41% amino acid sequence similarity with α-Klotho and is predominantly expressed in adipose tissue and the liver and pancreas. Functionally, β-Klotho acts as a cofactor for fibroblast growth factor 21 (FGF21), an endocrine factor synthesized in the liver that stimulates glucose uptake in adipocytes [158]. In this paper, the term Klotho will refer specifically to α-Klotho.
The human Klotho gene consists of five exons, which, through alternative RNA splicing, generate two distinct transcripts that encode either a membrane-bound or secreted protein. The first transcript is 3036 bp long (encoding 1012 amino acids) and encodes a single-pass membrane protein. This protein includes an N-terminal signal sequence, a putative extracellular domain with two internal repeats (KL1 and KL2, which share 21% similarity to each other), a single membrane-spanning region, and a short intracellular domain. The membrane protein can undergo enzymatic cleavage by ADAMTS 10/17, two members of the disintegrin and metalloprotease family, releasing the entire extracellular domain into the circulation [159,160].
The second transcript of Klotho includes the full 3036 bp encoding the membrane form, with an additional 50 bp insertion, bringing the total length to 3086 bp. However, this insertion contains a premature stop codon, which causes a block in the cDNA translation process, resulting in the synthesis of a truncated protein of 549 amino acids. This truncated protein retains the KL1 domain but lacks the second internal repeat of the extracellular domain (KL2), the transmembrane domain, and the intracellular domain. Consequently, this truncated protein is thought to be secreted outside the cell [161].
The existence of a secreted Klotho protein arising from this second transcript has not been definitively demonstrated. Moreover, recent advances in RNA surveillance suggest that the premature termination codons in the alternative Klotho mRNA (encoding the secreted form of Klotho) leads to mRNA degradation via nonsense-mediated decay (NMD) [162].
As a result, when the authors refer to the soluble form of Klotho (sKlotho) in this review, they are specifically referring to the form generated from the membrane protein (mKlotho) after enzymatic cleavage. mKlotho is predominantly expressed in normal kidneys, but also to a lesser extent in the brain, parathyroid glands, peripheral blood cells, vascular tissue, and placenta [47,163].
Interestingly, sKlotho is more abundant than mKlotho in several tissues, including the kidney, prostate, brain, hippocampus, placenta, and small intestine [161]. sKlotho is found in blood, urine, and cerebrospinal fluid [164] where it exerts cytoprotective effects on various organs, including the kidney. In the following section, the authors will discuss the protective roles of Klotho in the kidney, lung, brain, liver, and peripheral blood vessels (including cardiovascular diseases), which are key organs affected by dysfunctional disorders in age-related diseases and preeclamptic pregnancy.

6.1. Klotho Gene and Cytoprotective Effects

Due to its antioxidative properties, Klotho has been linked to several biological pathways and mechanisms. The first study investigating this property was published by Nagai et al. in 2003 [165] where the authors examined the role of oxidative stress in Klotho mutant mice. They focused on neurochemical and behavioral changes associated with aging and found that the level of 8-oxo-2′-deoxyguanosine (8-OHdG), a marker of oxidative DNA damage, was significantly elevated in the brains of these mutant mice. Interestingly, this increase was age-dependent. Additionally, the activities of antioxidant enzymes, such as Cu/Zn-SOD and GPx, were enhanced in the brains of the Klotho mutant mice. Two years later, Yamamoto et al. conducted a study that further confirmed Klotho’s protective role against oxidative stress, as suggested by Nagai et al. Their findings showed that Klotho-overexpressing mice lived longer, exhibited lower levels of 8-OHdG, and had higher levels of Mn-SOD compared to controls [166].
The protective effects of Klotho have also been observed in kidney diseases. In mice with immune-mediated glomerulonephritis, increasing Klotho levels through genetic manipulation improved kidney function and reduced mitochondrial oxidative stress, including DNA fragmentation, superoxide anion generation, lipid peroxidation, and apoptosis. In humans, low Klotho levels in blood, urine, and kidney have been strongly associated with acute kidney injury, with Klotho levels correlating with resilience to kidney damage [167]. In pregnancies complicated by preeclampsia, placental mKlotho expression, measured by real-time PCR, was found to be lower compared to healthy pregnancies [47,168]. Additionally, a significant downregulation of soluble Klotho (sKlotho) was observed in the urine of women with preeclampsia compared to healthy controls [64].
The sKlotho protein, released into circulation following enzymatic cleavage of mKlotho, exhibits cytoprotective effects that extend to distant organs, including the lung. In cases of acute kidney injury, blood circulation of sKlotho is reduced, which, in turn, increases the risk of pulmonary complications [169].
In A549 lung epithelial cells exposed to hyperoxic conditions (95% O2) and high inorganic phosphate concentrations (3–5 mM) to induce oxidative damage to DNA and proteins, Ravikumar et al. observed an increase in cellular antioxidant capacity when sKlotho-containing conditioned media was added. In vivo, they administered sKlotho-containing conditioned media into rat peritoneum before and during hyperoxia treatment, resulting in reduced alveolar interstitial edema and oxidative damage [155]. In elderly subjects with interstitial lung abnormalities, sKlotho levels in serum were found to be lower compared to healthy individuals, suggesting that sKlotho concentrations could serve as a predictive biomarker for accelerated lung function decline in individuals with interstitial lung abnormalities [170], although not for lung cancer [171].
The brain, being highly metabolically active, is particularly vulnerable to oxidative damage, which has been linked to several neurodegenerative diseases and mild cognitive impairment [172,173]. In 2014, Semba et al. studied 70 patients, both male and female, young and old, with and without Alzheimer’s disease. They measured sKlotho concentrations in cerebrospinal fluid using an ELISA assay. The results showed that sKlotho levels were lower in females compared to males (632–801 pg/mL vs. 814–983 pg/mL, p = 0.002), in patients with Alzheimer’s disease compared to those without (603–725 pg/mL vs. 705–828 pg/mL, p = 0.02), and in older adults compared to younger adults (658–874 pg/mL vs. 884–1100 pg/mL, p = 0.005) [174]. In a longitudinal study involving 1453 adults enrolled from 1998 to 2000, with a 3-year follow-up from 2001 to 2003, higher plasma sKlotho levels were found to be independently associated with better cognitive performance and less cognitive decline [175].
A study by Lim et al. was the first to characterize the tissue expression of mKlotho in humans. Immunohistochemistry revealed mKlotho expression in kidney, arterial, epithelial, endocrine, reproductive, and neuronal tissues, but not in the liver [176].
However, sKlotho also exerts a protective role in the liver by influencing its energy metabolism. Rao et al. investigated the effects of 5 weeks of peripheral administration of sKlotho in high-fat diet-induced obese mice. Their findings showed that sKlotho administration reduced lipid accumulation in both the liver and adipose tissue compared to controls. Though the exact mechanisms are not fully understood, this study highlights sKlotho’s potential role in regulating energy metabolism in adipose tissue [177], similar to the role of other hormones such as oxytocin [178].
The discovery that Klotho is also expressed in peripheral arterioles has led to the hypothesis that it may be linked to cardiovascular diseases [176]. Endothelial dysfunction is a key contributor to the development and progression of CVDs [179,180]. Nitric oxide (NO) plays a crucial role in maintaining vascular tone and endothelial function [181]. In endothelial cells, NO is synthesized from L-arginine by the enzyme nitric oxide synthase (NOS). Saito et al. first provided evidence that Klotho helps preserve endothelial function by stimulating NO production [182]. A subsequent study using animal models confirmed that Klotho deficiency in mice led to endothelial dysfunction, possibly due to reduced NO production [183]. In human aortic endothelial cells, endogenous Klotho promoted NO production following administration of FGF23 [184]. Reduced NO production, in turn, leads to compromised endothelial function, which is indicative of endothelial cell senescence.
Several studies suggest that Klotho’s role in endothelial cell senescence in CVDs is mediated by Sirtuin (SIRT1) [185]. SIRT1, highly expressed in endothelial cells, is involved in regulating aging-related processes such as endothelial cell senescence, inflammation, and apoptosis [186,187]. Research by Gao et al. demonstrated that Klotho-deficient mice with arterial stiffness and hypertension had reduced serum Klotho levels (approximately 45% lower) and that activating SIRT1 could reverse some of these effects. Their study also showed that Klotho deficiency led to decreased activity of endothelial nitric oxide synthase and AMP-activated protein kinase α (AMPKα) in aortic tissues [188]. In a similar vein, other studies have shown that supplementing with ghrelin, berberine, or exogenous Klotho can revert senescence in aging mice [189,190].

6.2. Klotho, Placenta and Preeclampsia

Klotho plays a crucial protective role in reproductive development. In pregnant mice, exposure to perfluorooctane sulfonate (PFOS) reduced Klotho expression in the placenta, increasing the risk of birth defects in fetuses [191]. Klotho knockout pigs failed to carry pregnancy to term [192]. Genetic studies in humans suggest that Klotho gene deficiency is associated with vascular calcification, premature bone disease, altered angiogenesis, hypertension, and left ventricular hypertrophy—hallmarks of premature vascular aging [193]. Research focusing on pregnancy has uncovered associations between Klotho and various obstetrical pathologies, particularly those related to placental aging and fetal growth. Recent studies have shown that pregnant women exhibit higher median plasma concentrations of α-Klotho compared to non-pregnant women, with levels increasing as pregnancy progresses. While the exact source of α-Klotho during pregnancy remains unclear, it is hypothesized that changes in maternal or placental production may contribute to its altered expression. It is believed that gene expression and protein synthesis predominantly occur in the placenta, rather than in the mother or fetus [194]. The synthesis of α-Klotho in the placenta is particularly noteworthy, as it plays key roles in angiogenesis [195], adipogenesis [196], calcium [197] and glucose metabolism [196], antioxidant effects [166], insulin signaling [196], and phosphate metabolism [198]. For instance, Ohata et al. reported that serum samples from the umbilical vein of newborns contained higher concentrations of α-Klotho compared to 4-day-old infants, mothers, and adult volunteers [194]. Similarly, Godang et al. found higher α-Klotho concentrations in neonatal umbilical cord plasma than in maternal plasma at 32–34 weeks of gestation, confirming its expression in the syncytiotrophoblast, the epithelial layer covering the highly vascular embryonic villi of the placenta, which is in direct contact with maternal blood [199]. Immunolocalization studies have shown that α-Klotho is predominantly located in the syncytiotrophoblast [194] along with ADAM17, a metallopeptidase acting as a TNFα-converting enzyme [200] and fibroblast growth factor receptor-1 (FGFR1) [201]. It is proposed that placental α-Klotho acts as a co-receptor for FGFR1 in the syncytiotrophoblast, and the complex of fibroblast growth factor 23 (FGF23), α-Klotho, and FGFR1 may contribute to vitamin D, phosphate, and calcium metabolism [201]. Furthermore, the shedding of α-Klotho by ADAM17 could release the protein into maternal circulation, potentially explaining the elevated plasma concentrations of α-Klotho during pregnancy [200]. Interestingly, ADAM17 levels are higher in placentas from pregnancies complicated by preeclampsia compared to those from normal pregnancies [202].
Placental development is a tightly regulated process during pregnancy, ensuring fetal growth by providing oxygen and nutrients while removing waste products such as carbon dioxide [37,203,204,205].
The placenta also acts as an endocrine organ, influencing both maternal and fetal metabolism. The median maternal plasma concentration of α-Klotho has been found to be lower in women with fetal growth retardation compared to those with normal-weight deliveries [206]. Shao et al. observed elevated α-Klotho mRNA and protein levels in the placentas of women with macrosomia, regardless of gestational diabetes, compared to those who delivered normal-weight babies [207]. Additionally, serum levels of α-Klotho in both maternal and umbilical cord blood were positively correlated with fetal birth weight [208], suggesting that α-Klotho plays a metabolic role in fetal growth. In contrast, lower maternal plasma levels of α-Klotho are observed in women who deliver small-for-gestational-age babies, independent of preeclampsia status [206]. The connection between Klotho and preeclampsia has been extensively studied, and Klotho is being considered as a potential marker for diagnosis and treatment [47,209]. However, the underlying mechanisms remain unclear. One possibility lies in the aging of the placenta. In normal pregnancies, oxidative stress during later stages is associated with an increase in syncytial nodes, which exhibit signs of oxidative damage, a characteristic of normal placental aging [210,211]. Syncytial node formation is further elevated in preeclampsia, suggesting premature placental aging. Despite expectations that α-Klotho, an anti-aging protein, would be linked to placental aging phenomena such as accelerated villous maturation, this association was not confirmed. Instead, maternal plasma levels of α-Klotho were higher in preeclampsia, despite the absence of accelerated villous maturation. This discrepancy has been explained by the finding that low maternal α-Klotho levels could activate the p53/p21 pathways, increasing cellular senescence and contributing to accelerated placental villous maturation [212,213]. In the context of placental oxidative stress, the Nrf2/ARE antioxidant signaling pathway plays a crucial role in maintaining oxidative balance and has been implicated in preeclampsia [9,214]. Klotho has been shown to modulate the Nrf2/ARE pathway [215], as it activates Nrf2 in podocytes to alleviate diabetic nephropathy [216]. Thus, Klotho may influence preeclampsia pathogenesis by modulating the Nrf2/ARE pathway. Studies also show that Klotho expression is downregulated in hypoxia-treated trophoblasts and preeclamptic rat tissues, suggesting a correlation between Klotho levels and disease progression [217]. Klotho is thought to exert its antioxidant effects through inhibition of the insulin/insulin-like growth factor-1 (IGF-1) pathway and activation of FoxO proteins. The next section will focus on the inhibition of the insulin/IGF-1 pathway and the activation of FoxO proteins.

6.3. Klotho Regulates Mytho Gene Expression via FoxO Proteins

The insulin/insulin-like growth factor-1 (IGF-1) signaling pathway plays a critical role in determining longevity, as demonstrated by various studies in organisms such as the nematode Caenorhabditis elegans, the fruit fly Drosophila melanogaster, and several rodent models [218].
This pathway involves IGF-1, its receptor (IGF-1R), and various binding proteins.
Insulin-like growth factor-1 (IGF-1) was first discovered in the late 1950s by researchers studying the mechanisms of growth hormone (GH, or somatotropin) in animal models [219]. The IGF-1 gene, located on chromosome 12, spans at least 90 kb of DNA and contains six exons [220]. Several IGF-1 mRNA variants have been identified, primarily due to alternative transcription initiation and splicing events [221]. These mRNAs differ mainly in their 5′ and 3′ untranslated regions [222]. The IGF-1 peptide is composed of 70 amino acids (~7.65 kDa) [223] and is released by the liver under the control of growth hormone [224].
The gene encoding the human IGF-1R is located on chromosome 15 and is ubiquitously expressed [225]. The receptor is a tetramer consisting of two extracellular α-chains and two intracellular β-chains [226]. The β-chains contain an intracellular tyrosine kinase domain, which undergoes autophosphorylation upon IGF-1 binding to IGF-1R [227]. This autophosphorylation activates the receptor, triggering the phosphorylation of several cellular proteins, including members of the IRS family [228]. Phosphorylated IRS proteins can activate phosphatidylinositol 3-kinase (PI3K) and Akt [229]. Akt is a serine/threonine kinase that has been extensively studied due to its involvement in various cellular processes including carcinogenesis [230,231]. One of the main targets of Akt activity is FoxO proteins [232]. Upon phosphorylation, FoxO proteins are retained in the cytoplasm and are unable to translocate to the nucleus to activate the transcription of their target genes [232].
FoxO proteins belong to a superfamily of forkhead transcription factors. Members of this superfamily share a conserved 110 amino acid DNA-binding domain known as the “forkhead” or “winged helix” domain [233]. The FoxO factors are homologous to DAF-16, a gene in Caenorhabditis elegans that plays a key role in regulating the aging process [234]. In mammals, there are four FoxO members: FoxO1, FoxO3, FoxO4, and FoxO6 [235]. In humans, FoxO1, FoxO3, and FoxO4 are ubiquitously expressed, including in placental tissue [236,237]. FoxO3 is particularly abundant in tissues such as the brain, heart, kidney, and spleen [238,239,240,241], while FoxO6 mRNA is predominantly expressed in the developing and adult brain, suggesting a role in the nervous system [242]. Several studies have shown that Klotho can inhibit the insulin/insulin-like growth factor-1 (IGF-1) signaling pathway [166,243]. By blocking the PI3K/Akt-mediated phosphorylation, FoxO factors translocate to the nucleus, where they promote the transcription of genes involved in metabolism, cell cycle regulation, apoptosis, stress resistance, DNA repair, and antioxidant defense. Key antioxidant genes such as catalase (CAT) and superoxide dismutase (SOD2) are among these targets [243,244,245]. Studies in animal models and human cell cultures have shown that Klotho overexpression or the addition of sKlotho to culture media increases Mn-SOD expression and reduces FoxO phosphorylation levels [166,246]. In the brain of senescence-accelerated mice (SAMP8), inhibition of the Akt/FoxO1 pathway led to increased expression of MnSOD and catalase, enhancing DNA repair [247]. In aged SAMP8 mice, Klotho deficiency was linked to FoxO1 inactivation and a reduction in antioxidant enzyme levels [247]. Further studies using Klotho +/− BALB/c mice demonstrated that administration of sKlotho dramatically reduced the level of 8-OHdG in urine, suggesting improved DNA repair activity through FoxO3a binding to the MnSOD promoter. This effect was dependent on Klotho-mediated inhibition of PI3K/Akt signaling [243].
Recently, Leduc-Gaudet and colleagues identified a new FoxO-dependent gene, Mytho (Macroautophagy and YouTH Optimizer) [248]. Though Mytho’s exact role is still being understood, experimental data suggest that it acts as a regulator of autophagy and skeletal muscle integrity. In mice with skeletal muscle atrophy, Mytho protein was overexpressed in muscle tissue, while silencing the Mytho gene resulted in increased muscle mass. Prolonged silencing led to severe myopathy. sKlotho may regulate Mytho expression through FoxO proteins (Figure 1). The prolonged silencing of Mytho may explain the musculoskeletal frailty observed in elderly individuals and women with preeclampsia.

7. Discussion

Pregnancy represents a unique biological state in a woman’s life and may serve as either a senescence or rejuvenation event for the body. Senescence plays a crucial role in various pregnancy-related processes, including syncytiotrophoblast formation, placental implantation, embryonic development, and delivery [249]. During pregnancy, many metabolic changes occur that contribute to aging, such as alterations in DNA methylation, increased inflammation, oxidative stress, and telomere shortening [250,251,252]. However, after a healthy pregnancy, women experience a period of rejuvenation and repair in the weeks and months following delivery [252]. It remains uncertain whether pregnancy accelerates, slows, or prolongs the aging process. The answer likely lies in the complexity of these events, which are influenced by a woman’s biological and genetic makeup, the type of pregnancy, and the postpartum timeline. Notably, pregnancy complications such as preeclampsia are associated with an increased risk of cardiovascular and neurovascular events in later life. These events tend to occur earlier in life than in women with uncomplicated pregnancies and are often preceded by a prolonged asymptomatic period.
The connection between preeclampsia and aging is supported by research on the Klotho gene, an anti-aging protein that appears to play a pivotal role during the intrauterine period and may influence long-term health outcomes. Current insights into the link between Klotho and intrauterine metabolism are promising, but need further exploration.

8. Conclusions

This review highlights the complex relationship between preeclampsia and aging, emphasizing shared biological and pathological features such as oxidative stress, inflammation, and cellular senescence. The Klotho gene emerges as a key regulator in these processes, exhibiting protective roles across various organ systems by modulating oxidative stress, inflammation, and metabolic pathways. Its involvement in placental function and pregnancy outcomes further underscores its importance in the pathogenesis of preeclampsia.
Future research should aim to elucidate the molecular mechanisms underlying Klotho’s actions and explore its potential as a biomarker for early diagnosis and therapeutic interventions. Furthermore, investigating the connection between Klotho expression and the recently identified Mytho gene offers a promising direction for understanding the broader implications of senescence and metabolic regulation in both pregnancy and aging.

9. Future Directions

The placenta plays a central role in preeclampsia, providing a unique lens through which to examine aging processes. Syncytial knots and other markers of premature placental aging are more prevalent in preeclampsia, reflecting oxidative stress and cellular senescence. The relationship between placental Klotho expression and fetal outcomes highlights its importance in intrauterine programming and long-term health. Notably, elevated levels of α-Klotho in maternal plasma during pregnancy may act as a compensatory response to counteract oxidative stress. However, its dysregulation in preeclampsia suggests potential diagnostic and therapeutic applications. Olivieri et al. demonstrated that long-term cultured HUVECs, a model of replicative cell senescence, release exosomes that induce a pro-senescent phenotype in co-cultured target cells [253]. Building on this, we propose the establishment of a co-culture system of long-term cultured HUVECs as a model for preeclampsia, with kidney or hepatic cells. Characterizing the exosomal content—such as miRNAs and proteins—could provide valuable insights into the potential role of Klotho as a clinical biomarker. This could open up new therapeutic avenues for managing pregnancy-related conditions and preventing fetal complications.
This review outlines several promising avenues for future research:
  • Mechanistic Studies: Further investigation into Klotho’s interactions with the Nrf2/ARE pathway, FoxO proteins, and epigenetic regulators could shed light on its protective mechanisms in both preeclampsia and aging.
  • Biomarker Development: Exploring Klotho as a biomarker for the early diagnosis of preeclampsia and age-related diseases is crucial. Profiling circulating Klotho levels, along with associated epigenetic markers and miRNAs, could enhance predictive accuracy.
  • Therapeutic Interventions: Developing strategies to upregulate Klotho expression or mimic its effects through pharmacological agents or gene therapy could revolutionize the management of preeclampsia and age-related disorders.

Author Contributions

Conceptualization, S.R.G., A.V. and M.C.; writing—original draft preparation, S.V., L.C., S.F. and M.C.; writing—review and editing, A.V. and S.R.G.; supervision, A.C. and M.E. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Abalos, E.; Cuesta, C.; Grosso, A.L.; Chou, D.; Say, L. Global and regional estimates of preeclampsia and eclampsia: A systematic review. Eur. J. Obstet. Gynecol. Reprod. Biol. 2013, 170, 1–7. [Google Scholar] [CrossRef]
  2. Steegers, E.A.; von Dadelszen, P.; Duvekot, J.J.; Pijnenborg, R. Pre-eclampsia. Lancet 2010, 376, 631–644. [Google Scholar] [CrossRef] [PubMed]
  3. ACOG Committee on Obstetric Practice. ACOG practice bulletin. Diagnosis and management of preeclampsia and eclampsia. Number 33, January 2002. Obstet. Gynecol. 2002, 99, 159–167. [Google Scholar] [CrossRef]
  4. Benschop, L.; Duvekot, J.J.; Versmissen, J.; van Broekhoven, V.; Steegers, E.A.P.; Roeters van Lennep, J.E. Blood Pressure Profile 1 Year after Severe Preeclampsia. Hypertension 2018, 71, 491–498. [Google Scholar] [CrossRef]
  5. Wu, P.; Haththotuwa, R.; Kwok, C.S.; Babu, A.; Kotronias, R.A.; Rushton, C.; Zaman, A.; Fryer, A.A.; Kadam, U.; Chew-Graham, C.A.; et al. Preeclampsia and Future Cardiovascular Health: A Systematic Review and Meta-Analysis. Circ. Cardiovasc. Qual. Outcomes 2017, 10, e003497. [Google Scholar] [CrossRef] [PubMed]
  6. Mongraw-Chaffin, M.L.; Cirillo, P.M.; Cohn, B.A. Preeclampsia and cardiovascular disease death: Prospective evidence from the child health and development studies cohort. Hypertension 2010, 56, 166–171. [Google Scholar] [CrossRef]
  7. Annesi, L.; Tossetta, G.; Borghi, C.; Piani, F. The Role of Xanthine Oxidase in Pregnancy Complications: A Systematic Review. Antioxidants 2024, 13, 1234. [Google Scholar] [CrossRef]
  8. Piani, F.; Tossetta, G.; Fantone, S.; Agostinis, C.; Di Simone, N.; Mandala, M.; Bulla, R.; Marzioni, D.; Borghi, C. First Trimester CD93 as a Novel Marker of Preeclampsia and Its Complications: A Pilot Study. High. Blood Press. Cardiovasc. Prev. 2023, 30, 591–594. [Google Scholar] [CrossRef]
  9. Tossetta, G.; Fantone, S.; Piani, F.; Crescimanno, C.; Ciavattini, A.; Giannubilo, S.R.; Marzioni, D. Modulation of NRF2/KEAP1 Signaling in Preeclampsia. Cells 2023, 12, 1545. [Google Scholar] [CrossRef] [PubMed]
  10. Piani, F.; Agnoletti, D.; Baracchi, A.; Scarduelli, S.; Verde, C.; Tossetta, G.; Montaguti, E.; Simonazzi, G.; Degli Esposti, D.; Borghi, C.; et al. Serum uric acid to creatinine ratio and risk of preeclampsia and adverse pregnancy outcomes. J. Hypertens. 2023, 41, 1333–1338. [Google Scholar] [CrossRef]
  11. Fantone, S.; Tossetta, G.; Di Simone, N.; Tersigni, C.; Scambia, G.; Marcheggiani, F.; Giannubilo, S.R.; Marzioni, D. CD93 a potential player in cytotrophoblast and endothelial cell migration. Cell Tissue Res. 2022, 387, 123–130. [Google Scholar] [CrossRef] [PubMed]
  12. Tossetta, G.; Fantone, S.; Gesuita, R.; Goteri, G.; Senzacqua, M.; Marcheggiani, F.; Tiano, L.; Marzioni, D.; Mazzucchelli, R. Ciliary Neurotrophic Factor Modulates Multiple Downstream Signaling Pathways in Prostate Cancer Inhibiting Cell Invasiveness. Cancers 2022, 14, 5917. [Google Scholar] [CrossRef] [PubMed]
  13. Tossetta, G.; Fantone, S.; Gesuita, R.; Di Renzo, G.C.; Meyyazhagan, A.; Tersigni, C.; Scambia, G.; Di Simone, N.; Marzioni, D. HtrA1 in Gestational Diabetes Mellitus: A Possible Biomarker? Diagnostics 2022, 12, 2705. [Google Scholar] [CrossRef]
  14. Tossetta, G.; Fantone, S.; Licini, C.; Marzioni, D.; Mattioli-Belmonte, M. The multifaced role of HtrA1 in the development of joint and skeletal disorders. Bone 2022, 157, 116350. [Google Scholar] [CrossRef] [PubMed]
  15. Cecati, M.; Sartini, D.; Campagna, R.; Biagini, A.; Ciavattini, A.; Emanuelli, M.; Giannubilo, S.R. Molecular analysis of endometrial inflammation in preterm birth. Cell Mol. Biol. 2017, 63, 51–57. [Google Scholar] [CrossRef]
  16. Marinelli Busilacchi, E.; Costantini, A.; Mancini, G.; Tossetta, G.; Olivieri, J.; Poloni, A.; Viola, N.; Butini, L.; Campanati, A.; Goteri, G.; et al. Nilotinib Treatment of Patients Affected by Chronic Graft-versus-Host Disease Reduces Collagen Production and Skin Fibrosis by Downmodulating the TGF-beta and p-SMAD Pathway. Biol. Blood Marrow Transplant. 2020, 26, 823–834. [Google Scholar] [CrossRef]
  17. Licini, C.; Tossetta, G.; Avellini, C.; Ciarmela, P.; Lorenzi, T.; Toti, P.; Gesuita, R.; Voltolini, C.; Petraglia, F.; Castellucci, M.; et al. Analysis of cell-cell junctions in human amnion and chorionic plate affected by chorioamnionitis. Histol. Histopathol. 2016, 31, 759–767. [Google Scholar] [CrossRef] [PubMed]
  18. Altobelli, E.; Latella, G.; Morroni, M.; Licini, C.; Tossetta, G.; Mazzucchelli, R.; Profeta, V.F.; Coletti, G.; Leocata, P.; Castellucci, M.; et al. Low HtrA1 expression in patients with long-standing ulcerative colitis and colorectal cancer. Oncol. Rep. 2017, 38, 418–426. [Google Scholar] [CrossRef]
  19. Harmon, A.C.; Cornelius, D.C.; Amaral, L.M.; Faulkner, J.L.; Cunningham, M.W., Jr.; Wallace, K.; LaMarca, B. The role of inflammation in the pathology of preeclampsia. Clin. Sci. 2016, 130, 409–419. [Google Scholar] [CrossRef]
  20. Piani, F.; Tossetta, G.; Cara-Fuentes, G.; Agnoletti, D.; Marzioni, D.; Borghi, C. Diagnostic and Prognostic Role of CD93 in Cardiovascular Disease: A Systematic Review. Biomolecules 2023, 13, 910. [Google Scholar] [CrossRef]
  21. Aouache, R.; Biquard, L.; Vaiman, D.; Miralles, F. Oxidative Stress in Preeclampsia and Placental Diseases. Int. J. Mol. Sci. 2018, 19, 1496. [Google Scholar] [CrossRef]
  22. Granger, J.P.; Spradley, F.T.; Bakrania, B.A. The Endothelin System: A Critical Player in the Pathophysiology of Preeclampsia. Curr. Hypertens. Rep. 2018, 20, 32. [Google Scholar] [CrossRef]
  23. Miller, E.C.; Wilczek, A.; Bello, N.A.; Tom, S.; Wapner, R.; Suh, Y. Pregnancy, preeclampsia and maternal aging: From epidemiology to functional genomics. Ageing Res. Rev. 2022, 73, 101535. [Google Scholar] [CrossRef] [PubMed]
  24. Cnattingius, S.; Reilly, M.; Pawitan, Y.; Lichtenstein, P. Maternal and fetal genetic factors account for most of familial aggregation of preeclampsia: A population-based Swedish cohort study. Am. J. Med. Genet. A 2004, 130A, 365–371. [Google Scholar] [CrossRef]
  25. Gray, K.J.; Saxena, R.; Karumanchi, S.A. Genetic predisposition to preeclampsia is conferred by fetal DNA variants near FLT1, a gene involved in the regulation of angiogenesis. Am. J. Obstet. Gynecol. 2018, 218, 211–218. [Google Scholar] [CrossRef]
  26. Steinthorsdottir, V.; McGinnis, R.; Williams, N.O.; Stefansdottir, L.; Thorleifsson, G.; Shooter, S.; Fadista, J.; Sigurdsson, J.K.; Auro, K.M.; Berezina, G.; et al. Genetic predisposition to hypertension is associated with preeclampsia in European and Central Asian women. Nat. Commun. 2020, 11, 5976. [Google Scholar] [CrossRef]
  27. Lisowska, M.; Pietrucha, T.; Sakowicz, A. Preeclampsia and Related Cardiovascular Risk: Common Genetic Background. Curr. Hypertens. Rep. 2018, 20, 71. [Google Scholar] [CrossRef]
  28. Tossetta, G.; Fantone, S.; Giannubilo, S.R.; Ciavattini, A.; Senzacqua, M.; Frontini, A.; Marzioni, D. HTRA1 in Placental Cell Models: A Possible Role in Preeclampsia. Curr. Issues Mol. Biol. 2023, 45, 3815–3828. [Google Scholar] [CrossRef] [PubMed]
  29. Tossetta, G.; Fantone, S.; Giannubilo, S.R.; Marinelli Busilacchi, E.; Ciavattini, A.; Castellucci, M.; Di Simone, N.; Mattioli-Belmonte, M.; Marzioni, D. Pre-eclampsia onset and SPARC: A possible involvement in placenta development. J. Cell Physiol. 2019, 234, 6091–6098. [Google Scholar] [CrossRef]
  30. Yang, F.; Janszky, I.; Gissler, M.; Roos, N.; Wikstrom, A.K.; Yu, Y.; Chen, H.; Bonamy, A.E.; Li, J.; Laszlo, K.D. Association of Maternal Preeclampsia With Offspring Risks of Ischemic Heart Disease and Stroke in Nordic Countries. JAMA Netw. Open 2022, 5, e2242064. [Google Scholar] [CrossRef]
  31. Huang, C.; Li, J.; Qin, G.; Liew, Z.; Hu, J.; Laszlo, K.D.; Tao, F.; Obel, C.; Olsen, J.; Yu, Y. Maternal hypertensive disorder of pregnancy and offspring early-onset cardiovascular disease in childhood, adolescence, and young adulthood: A national population-based cohort study. PLoS Med. 2021, 18, e1003805. [Google Scholar] [CrossRef]
  32. Rodgers, J.L.; Jones, J.; Bolleddu, S.I.; Vanthenapalli, S.; Rodgers, L.E.; Shah, K.; Karia, K.; Panguluri, S.K. Cardiovascular Risks Associated with Gender and Aging. J. Cardiovasc. Dev. Dis. 2019, 6, 19. [Google Scholar] [CrossRef]
  33. Lopez-Otin, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The hallmarks of aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef] [PubMed]
  34. Maynard, S.E.; Min, J.Y.; Merchan, J.; Lim, K.H.; Li, J.; Mondal, S.; Libermann, T.A.; Morgan, J.P.; Sellke, F.W.; Stillman, I.E.; et al. Excess placental soluble fms-like tyrosine kinase 1 (sFlt1) may contribute to endothelial dysfunction, hypertension, and proteinuria in preeclampsia. J. Clin. Investig. 2003, 111, 649–658. [Google Scholar] [CrossRef] [PubMed]
  35. Michalczyk, M.; Celewicz, A.; Celewicz, M.; Wozniakowska-Gondek, P.; Rzepka, R. The Role of Inflammation in the Pathogenesis of Preeclampsia. Mediators Inflamm. 2020, 2020, 3864941. [Google Scholar] [CrossRef]
  36. Fantone, S.; Mazzucchelli, R.; Giannubilo, S.R.; Ciavattini, A.; Marzioni, D.; Tossetta, G. AT-rich interactive domain 1A protein expression in normal and pathological pregnancies complicated by preeclampsia. Histochem. Cell Biol. 2020, 154, 339–346. [Google Scholar] [CrossRef]
  37. Fantone, S.; Giannubilo, S.R.; Marzioni, D.; Tossetta, G. HTRA family proteins in pregnancy outcome. Tissue Cell 2021, 72, 101549. [Google Scholar] [CrossRef]
  38. Lv, Z.; Xiong, L.L.; Qin, X.; Zhang, H.; Luo, X.; Peng, W.; Kilby, M.D.; Saffery, R.; Baker, P.N.; Qi, H.B. Role of GRK2 in Trophoblast Necroptosis and Spiral Artery Remodeling: Implications for Preeclampsia Pathogenesis. Front. Cell Dev. Biol. 2021, 9, 694261. [Google Scholar] [CrossRef]
  39. Seifer, D.B.; Lambert-Messerlian, G.; Palomaki, G.E.; Silver, R.M.; Parker, C.; Rowland Hogue, C.J.; Stoll, B.J.; Saade, G.R.; Goldenberg, R.L.; Dudley, D.J.; et al. Preeclampsia at delivery is associated with lower serum vitamin D and higher antiangiogenic factors: A case control study. Reprod. Biol. Endocrinol. 2022, 20, 8. [Google Scholar] [CrossRef]
  40. Ali, S.M.; Khalil, R.A. Genetic, immune and vasoactive factors in the vascular dysfunction associated with hypertension in pregnancy. Expert. Opin. Ther. Targets 2015, 19, 1495–1515. [Google Scholar] [CrossRef]
  41. van Niekerk, G.; Christowitz, C.; Engelbrecht, A.M. Insulin-mediated immune dysfunction in the development of preeclampsia. J. Mol. Med. 2021, 99, 889–897. [Google Scholar] [CrossRef] [PubMed]
  42. Leavey, K.; Benton, S.J.; Grynspan, D.; Kingdom, J.C.; Bainbridge, S.A.; Cox, B.J. Unsupervised Placental Gene Expression Profiling Identifies Clinically Relevant Subclasses of Human Preeclampsia. Hypertension 2016, 68, 137–147. [Google Scholar] [CrossRef]
  43. Hayflick, L.; Moorhead, P.S. The serial cultivation of human diploid cell strains. Exp. Cell Res. 1961, 25, 585–621. [Google Scholar] [CrossRef] [PubMed]
  44. Gorgoulis, V.; Adams, P.D.; Alimonti, A.; Bennett, D.C.; Bischof, O.; Bishop, C.; Campisi, J.; Collado, M.; Evangelou, K.; Ferbeyre, G.; et al. Cellular Senescence: Defining a Path Forward. Cell 2019, 179, 813–827. [Google Scholar] [CrossRef] [PubMed]
  45. Starr, J.M.; Shiels, P.G.; Harris, S.E.; Pattie, A.; Pearce, M.S.; Relton, C.L.; Deary, I.J. Oxidative stress, telomere length and biomarkers of physical aging in a cohort aged 79 years from the 1932 Scottish Mental Survey. Mech. Ageing Dev. 2008, 129, 745–751. [Google Scholar] [CrossRef]
  46. Rossiello, F.; Jurk, D.; Passos, J.F.; d’Adda di Fagagna, F. Telomere dysfunction in ageing and age-related diseases. Nat. Cell Biol. 2022, 24, 135–147. [Google Scholar] [CrossRef] [PubMed]
  47. Cecati, M.; Giannubilo, S.R.; Saccucci, F.; Sartini, D.; Ciavattini, A.; Emanuelli, M.; Tranquilli, A.L. Potential Role of Placental Klotho in the Pathogenesis of Preeclampsia. Cell Biochem. Biophys. 2016, 74, 49–57. [Google Scholar] [CrossRef]
  48. Xu, J.; Ye, J.; Wu, Y.; Zhang, H.; Luo, Q.; Han, C.; Ye, X.; Wang, H.; He, J.; Huang, H.; et al. Reduced fetal telomere length in gestational diabetes. PLoS ONE 2014, 9, e86161. [Google Scholar] [CrossRef]
  49. Biron-Shental, T.; Sukenik-Halevy, R.; Sharon, Y.; Goldberg-Bittman, L.; Kidron, D.; Fejgin, M.D.; Amiel, A. Short telomeres may play a role in placental dysfunction in preeclampsia and intrauterine growth restriction. Am. J. Obstet. Gynecol. 2010, 202, 381.e1–381.e7. [Google Scholar] [CrossRef]
  50. Hiyama, E.; Hiyama, K. Telomere and telomerase in stem cells. Br. J. Cancer 2007, 96, 1020–1024. [Google Scholar] [CrossRef]
  51. Kirstetter, P.; Anderson, K.; Porse, B.T.; Jacobsen, S.E.; Nerlov, C. Activation of the canonical Wnt pathway leads to loss of hematopoietic stem cell repopulation and multilineage differentiation block. Nat. Immunol. 2006, 7, 1048–1056. [Google Scholar] [CrossRef] [PubMed]
  52. Ahmed, A.S.; Sheng, M.H.; Wasnik, S.; Baylink, D.J.; Lau, K.W. Effect of aging on stem cells. World J. Exp. Med. 2017, 7, 1–10. [Google Scholar] [CrossRef] [PubMed]
  53. Moiseeva, V.; Cisneros, A.; Sica, V.; Deryagin, O.; Lai, Y.; Jung, S.; Andres, E.; An, J.; Segales, J.; Ortet, L.; et al. Author Correction: Senescence atlas reveals an aged-like inflamed niche that blunts muscle regeneration. Nature 2023, 614, E45. [Google Scholar] [CrossRef]
  54. Rando, T.A. Stem cells, ageing and the quest for immortality. Nature 2006, 441, 1080–1086. [Google Scholar] [CrossRef] [PubMed]
  55. Tchkonia, T.; Zhu, Y.; van Deursen, J.; Campisi, J.; Kirkland, J.L. Cellular senescence and the senescent secretory phenotype: Therapeutic opportunities. J. Clin. Investig. 2013, 123, 966–972. [Google Scholar] [CrossRef] [PubMed]
  56. Schafer, M.J.; Zhang, X.; Kumar, A.; Atkinson, E.J.; Zhu, Y.; Jachim, S.; Mazula, D.L.; Brown, A.K.; Berning, M.; Aversa, Z.; et al. The senescence-associated secretome as an indicator of age and medical risk. JCI Insight 2020, 5, 133668. [Google Scholar] [CrossRef]
  57. Ferrucci, L.; Corsi, A.; Lauretani, F.; Bandinelli, S.; Bartali, B.; Taub, D.D.; Guralnik, J.M.; Longo, D.L. The origins of age-related proinflammatory state. Blood 2005, 105, 2294–2299. [Google Scholar] [CrossRef] [PubMed]
  58. Wieczorowska-Tobis, K.; Niemir, Z.I.; Podkowka, R.; Korybalska, K.; Mossakowska, M.; Breborowicz, A. Can an increased level of circulating IL-8 be a predictor of human longevity? Med. Sci. Monit. 2006, 12, CR118-121. [Google Scholar]
  59. Eren, M.; Boe, A.E.; Klyachko, E.A.; Vaughan, D.E. Role of plasminogen activator inhibitor-1 in senescence and aging. Semin. Thromb. Hemost. 2014, 40, 645–651. [Google Scholar] [CrossRef]
  60. Brenta, G.; Nepote, A.; Barreto, A.; Musso, C.; Faingold, C.; Fossati, P.; Antonelli, A.; Fallahi, P.; Fama, F.; Merono, T. Low glomerular filtration rate values are associated with higher TSH in an elderly population at high cardiovascular disease risk. Front. Endocrinol. 2023, 14, 1162626. [Google Scholar] [CrossRef]
  61. Zimmermann, H.W.; Seidler, S.; Gassler, N.; Nattermann, J.; Luedde, T.; Trautwein, C.; Tacke, F. Interleukin-8 is activated in patients with chronic liver diseases and associated with hepatic macrophage accumulation in human liver fibrosis. PLoS ONE 2011, 6, e21381. [Google Scholar] [CrossRef]
  62. Duisenbek, A.; Aviles Perez, M.D.; Perez, M.; Aguilar Benitez, J.M.; Pereira Perez, V.R.; Gorts Ortega, J.; Ussipbek, B.; Yessenbekova, A.; Lopez-Armas, G.C.; Ablaikhanova, N.; et al. Unveiling the Predictive Model for Macrovascular Complications in Type 2 Diabetes Mellitus: microRNAs Expression, Lipid Profile, and Oxidative Stress Markers. Int. J. Mol. Sci. 2024, 25, 1763. [Google Scholar] [CrossRef] [PubMed]
  63. Stubblefield, W.B.; Alves, N.J.; Rondina, M.T.; Kline, J.A. Variable Resistance to Plasminogen Activator Initiated Fibrinolysis for Intermediate-Risk Pulmonary Embolism. PLoS ONE 2016, 11, e0148747. [Google Scholar] [CrossRef]
  64. Suvakov, S.; Ghamrawi, R.; Cubro, H.; Tu, H.; White, W.M.; Tobah, Y.S.B.; Milic, N.M.; Grande, J.P.; Cunningham, J.M.; Chebib, F.T.; et al. Epigenetic and senescence markers indicate an accelerated ageing-like state in women with preeclamptic pregnancies. EBioMedicine 2021, 70, 103536. [Google Scholar] [CrossRef] [PubMed]
  65. Macedo, A.P.A.; da Silva, A.S.R.; Munoz, V.R.; Ropelle, E.R.; Pauli, J.R. Mitochondrial dysfunction plays an essential role in remodeling aging adipose tissue. Mech. Ageing Dev. 2021, 200, 111598. [Google Scholar] [CrossRef] [PubMed]
  66. Bakula, D.; Scheibye-Knudsen, M. MitophAging: Mitophagy in Aging and Disease. Front. Cell Dev. Biol. 2020, 8, 239. [Google Scholar] [CrossRef] [PubMed]
  67. Juan, C.A.; Perez de la Lastra, J.M.; Plou, F.J.; Perez-Lebena, E. The Chemistry of Reactive Oxygen Species (ROS) Revisited: Outlining Their Role in Biological Macromolecules (DNA, Lipids and Proteins) and Induced Pathologies. Int. J. Mol. Sci. 2021, 22, 4642. [Google Scholar] [CrossRef] [PubMed]
  68. Zampino, M.; Spencer, R.G.; Fishbein, K.W.; Simonsick, E.M.; Ferrucci, L. Cardiovascular Health and Mitochondrial Function: Testing an Association. J. Gerontol. A Biol. Sci. Med. Sci. 2021, 76, 361–367. [Google Scholar] [CrossRef]
  69. Ashar, F.N.; Zhang, Y.; Longchamps, R.J.; Lane, J.; Moes, A.; Grove, M.L.; Mychaleckyj, J.C.; Taylor, K.D.; Coresh, J.; Rotter, J.I.; et al. Association of Mitochondrial DNA Copy Number With Cardiovascular Disease. JAMA Cardiol. 2017, 2, 1247–1255. [Google Scholar] [CrossRef]
  70. Kim, J.H.; Im, J.A.; Lee, D.C. The relationship between leukocyte mitochondrial DNA contents and metabolic syndrome in postmenopausal women. Menopause 2012, 19, 582–587. [Google Scholar] [CrossRef]
  71. Byappanahalli, A.M.; Noren Hooten, N.; Vannoy, M.; Mode, N.A.; Ezike, N.; Zonderman, A.B.; Evans, M.K. Mitochondrial DNA and inflammatory proteins are higher in extracellular vesicles from frail individuals. Immun. Ageing 2023, 20, 6. [Google Scholar] [CrossRef] [PubMed]
  72. Berkowitz, K.; Monteagudo, A.; Marks, F.; Jackson, U.; Baxi, L. Mitochondrial myopathy and preeclampsia associated with pregnancy. Am. J. Obstet. Gynecol. 1990, 162, 146–147. [Google Scholar] [CrossRef] [PubMed]
  73. Torbergsen, T.; Oian, P.; Mathiesen, E.; Borud, O. Pre-eclampsia--a mitochondrial disease? Acta Obstet. Gynecol. Scand. 1989, 68, 145–148. [Google Scholar] [CrossRef]
  74. Myatt, L.; Muralimanoharan, S.; Maloyan, A. Effect of preeclampsia on placental function: Influence of sexual dimorphism, microRNA’s and mitochondria. Adv. Exp. Med. Biol. 2014, 814, 133–146. [Google Scholar] [CrossRef]
  75. Marschalek, J.; Wohlrab, P.; Ott, J.; Wojta, J.; Speidl, W.; Klein, K.U.; Kiss, H.; Pateisky, P.; Zeisler, H.; Kuessel, L. Maternal serum mitochondrial DNA (mtDNA) levels are elevated in preeclampsia—A matched case-control study. Pregnancy Hypertens. 2018, 14, 195–199. [Google Scholar] [CrossRef] [PubMed]
  76. Waddington, C.H. Towards a theoretical biology. Nature 1968, 218, 525–527. [Google Scholar] [CrossRef] [PubMed]
  77. Wu, C.; Morris, J.R. Genes, genetics, and epigenetics: A correspondence. Science 2001, 293, 1103–1105. [Google Scholar] [CrossRef] [PubMed]
  78. Robertson, K.D. DNA methylation and human disease. Nat. Rev. Genet. 2005, 6, 597–610. [Google Scholar] [CrossRef] [PubMed]
  79. Pozzi, V.; Campagna, R.; Sartini, D.; Emanuelli, M. Nicotinamide N-Methyltransferase as Promising Tool for Management of Gastrointestinal Neoplasms. Biomolecules 2022, 12, 1173. [Google Scholar] [CrossRef]
  80. Jin, B.; Li, Y.; Robertson, K.D. DNA methylation: Superior or subordinate in the epigenetic hierarchy? Genes. Cancer 2011, 2, 607–617. [Google Scholar] [CrossRef]
  81. Bestor, T.H. The DNA methyltransferases of mammals. Hum. Mol. Genet. 2000, 9, 2395–2402. [Google Scholar] [CrossRef] [PubMed]
  82. White, W.M.; Brost, B.; Sun, Z.; Rose, C.; Craici, I.; Wagner, S.J.; Turner, S.T.; Garovic, V.D. Genome-wide methylation profiling demonstrates hypermethylation in maternal leukocyte DNA in preeclamptic compared to normotensive pregnancies. Hypertens. Pregnancy 2013, 32, 257–269. [Google Scholar] [CrossRef] [PubMed]
  83. Rahat, B.; Thakur, S.; Bagga, R.; Kaur, J. Epigenetic regulation of STAT5A and its role as fetal DNA epigenetic marker during placental development and dysfunction. Placenta 2016, 44, 46–53. [Google Scholar] [CrossRef]
  84. Zakeri, S.; Rahimi, Z.; Rezvani, N.; Vaisi-Raygani, A.; Alibakhshi, R.; Zakeri, S.; Yari, K. The influence of Nrf2 gene promoter methylation on gene expression and oxidative stress parameters in preeclampsia. BMC Med. Genomics 2024, 17, 64. [Google Scholar] [CrossRef]
  85. Harati-Sadegh, M.; Kohan, L.; Teimoori, B.; Mehrabani, M.; Salimi, S. Analysis of polymorphisms, promoter methylation, and mRNA expression profile of maternal and placental P53 and P21 genes in preeclamptic and normotensive pregnant women. J. Biomed. Sci. 2019, 26, 92. [Google Scholar] [CrossRef]
  86. Ye, W.; Shen, L.; Xiong, Y.; Zhou, Y.; Gu, H.; Yang, Z. Preeclampsia is Associated with Decreased Methylation of the GNA12 Promoter. Ann. Hum. Genet. 2016, 80, 7–10. [Google Scholar] [CrossRef]
  87. Zhang, L.; Zhao, F.; Yang, C.; Tang, Q.; Zhang, R.; Li, J.; Chen, A.; Hou, L.; Liu, S. Hypomethylation of CTGF Promoter in Placenta and Peripheral Blood of Pre-eclampsia Women. Reprod. Sci. 2020, 27, 468–476. [Google Scholar] [CrossRef]
  88. Mousa, A.A.; Strauss, J.F., 3rd; Walsh, S.W. Reduced methylation of the thromboxane synthase gene is correlated with its increased vascular expression in preeclampsia. Hypertension 2012, 59, 1249–1255. [Google Scholar] [CrossRef]
  89. Mousa, A.A.; Cappello, R.E.; Estrada-Gutierrez, G.; Shukla, J.; Romero, R.; Strauss, J.F., 3rd; Walsh, S.W. Preeclampsia is associated with alterations in DNA methylation of genes involved in collagen metabolism. Am. J. Pathol. 2012, 181, 1455–1463. [Google Scholar] [CrossRef] [PubMed]
  90. Walsh, S.W.; Nugent, W.H.; Archer, K.J.; Al Dulaimi, M.; Washington, S.L.; Strauss, J.F., 3rd. Epigenetic Regulation of Interleukin-17-Related Genes and Their Potential Roles in Neutrophil Vascular Infiltration in Preeclampsia. Reprod. Sci. 2022, 29, 154–162. [Google Scholar] [CrossRef]
  91. Roman-Gomez, J.; Castillejo, J.A.; Jimenez, A.; Gonzalez, M.G.; Moreno, F.; Rodriguez Mdel, C.; Barrios, M.; Maldonado, J.; Torres, A. 5′ CpG island hypermethylation is associated with transcriptional silencing of the p21(CIP1/WAF1/SDI1) gene and confers poor prognosis in acute lymphoblastic leukemia. Blood 2002, 99, 2291–2296. [Google Scholar] [CrossRef] [PubMed]
  92. Zhang, H.; Cai, X.; Yi, B.; Huang, J.; Wang, J.; Sun, J. Correlation of CTGF gene promoter methylation with CTGF expression in type 2 diabetes mellitus with or without nephropathy. Mol. Med. Rep. 2014, 9, 2138–2144. [Google Scholar] [CrossRef] [PubMed]
  93. Wei, L.; Liu, B.; Tuo, J.; Shen, D.; Chen, P.; Li, Z.; Liu, X.; Ni, J.; Dagur, P.; Sen, H.N.; et al. Hypomethylation of the IL17RC promoter associates with age-related macular degeneration. Cell Rep. 2012, 2, 1151–1158. [Google Scholar] [CrossRef]
  94. Jin, B.; Robertson, K.D. DNA methyltransferases, DNA damage repair, and cancer. Adv. Exp. Med. Biol. 2013, 754, 3–29. [Google Scholar] [CrossRef] [PubMed]
  95. Winter, J.; Jung, S.; Keller, S.; Gregory, R.I.; Diederichs, S. Many roads to maturity: microRNA biogenesis pathways and their regulation. Nat. Cell Biol. 2009, 11, 228–234. [Google Scholar] [CrossRef] [PubMed]
  96. Mayya, V.K.; Duchaine, T.F. Ciphers and Executioners: How 3′-Untranslated Regions Determine the Fate of Messenger RNAs. Front. Genet. 2019, 10, 6. [Google Scholar] [CrossRef]
  97. Broughton, J.P.; Lovci, M.T.; Huang, J.L.; Yeo, G.W.; Pasquinelli, A.E. Pairing beyond the Seed Supports MicroRNA Targeting Specificity. Mol. Cell 2016, 64, 320–333. [Google Scholar] [CrossRef] [PubMed]
  98. Arroyo, J.D.; Chevillet, J.R.; Kroh, E.M.; Ruf, I.K.; Pritchard, C.C.; Gibson, D.F.; Mitchell, P.S.; Bennett, C.F.; Pogosova-Agadjanyan, E.L.; Stirewalt, D.L.; et al. Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. Proc. Natl. Acad. Sci. USA 2011, 108, 5003–5008. [Google Scholar] [CrossRef]
  99. Wang, K.; Zhang, S.; Weber, J.; Baxter, D.; Galas, D.J. Export of microRNAs and microRNA-protective protein by mammalian cells. Nucleic Acids Res. 2010, 38, 7248–7259. [Google Scholar] [CrossRef] [PubMed]
  100. Chen, X.; Ba, Y.; Ma, L.; Cai, X.; Yin, Y.; Wang, K.; Guo, J.; Zhang, Y.; Chen, J.; Guo, X.; et al. Characterization of microRNAs in serum: A novel class of biomarkers for diagnosis of cancer and other diseases. Cell Res. 2008, 18, 997–1006. [Google Scholar] [CrossRef] [PubMed]
  101. Turchinovich, A.; Weiz, L.; Langheinz, A.; Burwinkel, B. Characterization of extracellular circulating microRNA. Nucleic Acids Res. 2011, 39, 7223–7233. [Google Scholar] [CrossRef]
  102. Giannubilo, S.R.; Cecati, M.; Marzioni, D.; Ciavattini, A. Circulating miRNAs and Preeclampsia: From Implantation to Epigenetics. Int. J. Mol. Sci. 2024, 25, 1418. [Google Scholar] [CrossRef]
  103. Tessier, N.P.; Hardy, L.M.; Deleuze, J.F.; How-Kit, A. Circulating cell-free nucleic acids of plasma in human aging, healthy aging and longevity: Current state of knowledge. Front. Genet. 2023, 14, 1321280. [Google Scholar] [CrossRef]
  104. Li, Q.; Han, Y.; Xu, P.; Yin, L.; Si, Y.; Zhang, C.; Meng, Y.; Feng, W.; Pan, Z.; Gao, Z.; et al. Elevated microRNA-125b inhibits cytotrophoblast invasion and impairs endothelial cell function in preeclampsia. Cell Death Discov. 2020, 6, 35. [Google Scholar] [CrossRef] [PubMed]
  105. Winger, E.E.; Reed, J.L.; Ji, X. First-trimester maternal cell microRNA is a superior pregnancy marker to immunological testing for predicting adverse pregnancy outcome. J. Reprod. Immunol. 2015, 110, 22–35. [Google Scholar] [CrossRef] [PubMed]
  106. Martinez-Fierro, M.L.; Garza-Veloz, I. Analysis of Circulating microRNA Signatures and Preeclampsia Development. Cells 2021, 10, 1003. [Google Scholar] [CrossRef]
  107. Jairajpuri, D.S.; Malalla, Z.H.; Mahmood, N.; Almawi, W.Y. Circulating microRNA expression as predictor of preeclampsia and its severity. Gene 2017, 627, 543–548. [Google Scholar] [CrossRef]
  108. Ye, Z.; Sun, B.; Mi, X.; Xiao, Z. Gene co-expression network for analysis of plasma exosomal miRNAs in the elderly as markers of aging and cognitive decline. PeerJ 2020, 8, e8318. [Google Scholar] [CrossRef]
  109. Mengel-From, J.; Feddersen, S.; Halekoh, U.; Heegaard, N.H.H.; McGue, M.; Christensen, K.; Tan, Q.; Christiansen, L. Circulating microRNAs disclose biology of normal cognitive function in healthy elderly people—A discovery twin study. Eur. J. Hum. Genet. 2018, 26, 1378–1387. [Google Scholar] [CrossRef] [PubMed]
  110. Sheinerman, K.S.; Tsivinsky, V.G.; Abdullah, L.; Crawford, F.; Umansky, S.R. Plasma microRNA biomarkers for detection of mild cognitive impairment: Biomarker validation study. Aging 2013, 5, 925–938. [Google Scholar] [CrossRef]
  111. Kocijan, R.; Muschitz, C.; Geiger, E.; Skalicky, S.; Baierl, A.; Dormann, R.; Plachel, F.; Feichtinger, X.; Heimel, P.; Fahrleitner-Pammer, A.; et al. Circulating microRNA Signatures in Patients With Idiopathic and Postmenopausal Osteoporosis and Fragility Fractures. J. Clin. Endocrinol. Metab. 2016, 101, 4125–4134. [Google Scholar] [CrossRef]
  112. Marchegiani, F.; Recchioni, R.; Di Rosa, M.; Piacenza, F.; Marcheselli, F.; Bonfigli, A.R.; Galeazzi, R.; Matacchione, G.; Cardelli, M.; Procopio, A.D.; et al. Low circulating levels of miR-17 and miR-126-3p are associated with increased mortality risk in geriatric hospitalized patients affected by cardiovascular multimorbidity. Geroscience 2024, 46, 2531–2544. [Google Scholar] [CrossRef]
  113. Olivieri, F.; Spazzafumo, L.; Santini, G.; Lazzarini, R.; Albertini, M.C.; Rippo, M.R.; Galeazzi, R.; Abbatecola, A.M.; Marcheselli, F.; Monti, D.; et al. Age-related differences in the expression of circulating microRNAs: miR-21 as a new circulating marker of inflammaging. Mech. Ageing Dev. 2012, 133, 675–685. [Google Scholar] [CrossRef]
  114. Gao, T.; Luo, J.; Fan, J.; Gong, G.; Yang, H. Epigenetic modifications associated to diabetic peripheral neuropathic pain (Review). Mol. Med. Rep. 2025, 31, 13394. [Google Scholar] [CrossRef]
  115. Pozzi, V.; Campagna, R.; Sartini, D.; Emanuelli, M. Enzymes Dysregulation in Cancer: From Diagnosis to Therapeutical Approaches. Int. J. Mol. Sci. 2023, 24, 13815. [Google Scholar] [CrossRef] [PubMed]
  116. Sterner, D.E.; Berger, S.L. Acetylation of histones and transcription-related factors. Microbiol. Mol. Biol. Rev. 2000, 64, 435–459. [Google Scholar] [CrossRef]
  117. Gibney, E.R.; Nolan, C.M. Epigenetics and gene expression. Heredity 2010, 105, 4–13. [Google Scholar] [CrossRef]
  118. Zhang, B.; Kim, M.Y.; Elliot, G.; Zhou, Y.; Zhao, G.; Li, D.; Lowdon, R.F.; Gormley, M.; Kapidzic, M.; Robinson, J.F.; et al. Human placental cytotrophoblast epigenome dynamics over gestation and alterations in placental disease. Dev. Cell 2021, 56, 1238–1252 e1235. [Google Scholar] [CrossRef] [PubMed]
  119. Meister, S.; Hahn, L.; Beyer, S.; Kuhn, C.; Jegen, M.; von Schonfeldt, V.; Corradini, S.; Schulz, C.; Kolben, T.M.; Hester, A.; et al. Epigenetic modification via H3K4me3 and H3K9ac in human placenta is reduced in preeclampsia. J. Reprod. Immunol. 2021, 145, 103287. [Google Scholar] [CrossRef]
  120. Sheng, W.; Gu, Y.; Chu, X.; Morgan, J.A.; Cooper, D.B.; Lewis, D.F.; McCathran, C.E.; Wang, Y. Upregulation of histone H3K9 methylation in fetal endothelial cells from preeclamptic pregnancies. J. Cell Physiol. 2021, 236, 1866–1874. [Google Scholar] [CrossRef]
  121. Whongsiri, P.; Pimratana, C.; Wijitsettakul, U.; Sanpavat, A.; Jindatip, D.; Hoffmann, M.J.; Goering, W.; Schulz, W.A.; Boonla, C. Oxidative stress and LINE-1 reactivation in bladder cancer are epigenetically linked through active chromatin formation. Free Radic. Biol. Med. 2019, 134, 419–428. [Google Scholar] [CrossRef]
  122. Kang, J.; Benjamin, D.I.; Kim, S.; Salvi, J.S.; Dhaliwal, G.; Lam, R.; Goshayeshi, A.; Brett, J.O.; Liu, L.; Rando, T.A. Depletion of SAM leading to loss of heterochromatin drives muscle stem cell ageing. Nat. Metab. 2024, 6, 153–168. [Google Scholar] [CrossRef] [PubMed]
  123. Chen, C.; Zhao, M.; Yin, N.; He, B.; Wang, B.; Yuan, Y.; Yu, F.; Hu, J.; Yin, B.; Lu, Q. Abnormal histone acetylation and methylation levels in esophageal squamous cell carcinomas. Cancer Investig. 2011, 29, 548–556. [Google Scholar] [CrossRef]
  124. Onyango, I.G.; Bennett, J.P.; Stokin, G.B. Mitochondrially-Targeted Therapeutic Strategies for Alzheimer’s Disease. Curr. Alzheimer Res. 2021, 18, 753–771. [Google Scholar] [CrossRef]
  125. Tain, Y.L.; Hsu, C.N. Interplay between maternal nutrition and epigenetic programming on offspring hypertension. J. Nutr. Biochem. 2024, 127, 109604. [Google Scholar] [CrossRef]
  126. Emanuelli, M.; Sartini, D.; Molinelli, E.; Campagna, R.; Pozzi, V.; Salvolini, E.; Simonetti, O.; Campanati, A.; Offidani, A. The Double-Edged Sword of Oxidative Stress in Skin Damage and Melanoma: From Physiopathology to Therapeutical Approaches. Antioxidants 2022, 11, 612. [Google Scholar] [CrossRef] [PubMed]
  127. Birben, E.; Sahiner, U.M.; Sackesen, C.; Erzurum, S.; Kalayci, O. Oxidative stress and antioxidant defense. World Allergy Organ. J. 2012, 5, 9–19. [Google Scholar] [CrossRef] [PubMed]
  128. Campagna, R.; Pozzi, V.; Giorgini, S.; Morichetti, D.; Goteri, G.; Sartini, D.; Serritelli, E.N.; Emanuelli, M. Paraoxonase-2 is upregulated in triple negative breast cancer and contributes to tumor progression and chemoresistance. Hum. Cell 2023, 36, 1108–1119. [Google Scholar] [CrossRef]
  129. Bacchetti, T.; Campagna, R.; Sartini, D.; Cecati, M.; Morresi, C.; Bellachioma, L.; Martinelli, E.; Rocchetti, G.; Lucini, L.; Ferretti, G.; et al. C. spinosa L. subsp. rupestris Phytochemical Profile and Effect on Oxidative Stress in Normal and Cancer Cells. Molecules 2022, 27, 6488. [Google Scholar] [CrossRef] [PubMed]
  130. Milkovic, L.; Cipak Gasparovic, A.; Cindric, M.; Mouthuy, P.A.; Zarkovic, N. Short Overview of ROS as Cell Function Regulators and Their Implications in Therapy Concepts. Cells 2019, 8, 793. [Google Scholar] [CrossRef] [PubMed]
  131. Roy, J.; Galano, J.M.; Durand, T.; Le Guennec, J.Y.; Lee, J.C. Physiological role of reactive oxygen species as promoters of natural defenses. FASEB J. 2017, 31, 3729–3745. [Google Scholar] [CrossRef] [PubMed]
  132. Gusar, V.A.; Timofeeva, A.V.; Chagovets, V.V.; Vysokikh, M.Y.; Kan, N.E.; Manukhova, L.A.; Marey, M.V.; Sukhikh, G.T. Interrelation between miRNAs Expression Associated with Redox State Fluctuations, Immune and Inflammatory Response Activation, and Neonatal Outcomes in Complicated Pregnancy, Accompanied by Placental Insufficiency. Antioxidants 2022, 12, 6. [Google Scholar] [CrossRef]
  133. Cecati, M.; Emanuelli, M.; Giannubilo, S.R.; Quarona, V.; Senetta, R.; Malavasi, F.; Tranquilli, A.L.; Saccucci, F. Contribution of adenosine-producing ectoenzymes to the mechanisms underlying the mitigation of maternal-fetal conflicts. J. Biol. Regul. Homeost. Agents 2013, 27, 519–529. [Google Scholar] [PubMed]
  134. Delli Muti, N.; Salvio, G.; Ciarloni, A.; Perrone, M.; Tossetta, G.; Lazzarini, R.; Bracci, M.; Balercia, G. Can extremely low frequency magnetic field affect human sperm parameters and male fertility? Tissue Cell 2023, 82, 102045. [Google Scholar] [CrossRef]
  135. Tossetta, G.; Fantone, S.; Marzioni, D.; Mazzucchelli, R. Role of Natural and Synthetic Compounds in Modulating NRF2/KEAP1 Signaling Pathway in Prostate Cancer. Cancers 2023, 15, 3037. [Google Scholar] [CrossRef]
  136. Fantone, S.; Tossetta, G.; Cianfruglia, L.; Frontini, A.; Armeni, T.; Procopio, A.D.; Pugnaloni, A.; Gualtieri, A.F.; Marzioni, D. Mechanisms of action of mineral fibres in a placental syncytiotrophoblast model: An in vitro toxicology study. Chem. Biol. Interact. 2024, 390, 110895. [Google Scholar] [CrossRef] [PubMed]
  137. Tossetta, G.; Fantone, S.; Togni, L.; Santarelli, A.; Olivieri, F.; Marzioni, D.; Rippo, M.R. Modulation of NRF2/KEAP1 Signaling by Phytotherapeutics in Periodontitis. Antioxidants 2024, 13, 1270. [Google Scholar] [CrossRef]
  138. Fantone, S.; Marzioni, D.; Tossetta, G. NRF2/KEAP1 signaling inhibitors in gynecologic cancers. Expert. Rev. Anticancer. Ther. 2024, 24, 1191–1194. [Google Scholar] [CrossRef] [PubMed]
  139. Link, W.; Ferreira, B.I. FOXO Transcription Factors: A Brief Overview. Methods Mol. Biol. 2025, 2871, 1–8. [Google Scholar] [CrossRef]
  140. Solcia, E.; Villani, L.; Luinetti, O.; Fiocca, R. Proton pump inhibitors, enterochromaffin-like cell growth and Helicobacter pylori gastritis. Aliment. Pharmacol. Ther. 1993, 7 (Suppl. S1), 25–28, discussion 29–31. [Google Scholar] [CrossRef]
  141. Campbell, E.L.; Colgan, S.P. Control and dysregulation of redox signalling in the gastrointestinal tract. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 106–120. [Google Scholar] [CrossRef]
  142. Campagna, R.; Belloni, A.; Pozzi, V.; Salvucci, A.; Notarstefano, V.; Togni, L.; Mascitti, M.; Sartini, D.; Giorgini, E.; Salvolini, E.; et al. Role Played by Paraoxonase-2 Enzyme in Cell Viability, Proliferation and Sensitivity to Chemotherapy of Oral Squamous Cell Carcinoma Cell Lines. Int. J. Mol. Sci. 2022, 24, 338. [Google Scholar] [CrossRef] [PubMed]
  143. Sultana, Z.; Maiti, K.; Aitken, J.; Morris, J.; Dedman, L.; Smith, R. Oxidative stress, placental ageing-related pathologies and adverse pregnancy outcomes. Am. J. Reprod. Immunol. 2017, 77, e12653. [Google Scholar] [CrossRef]
  144. Roberts, J.M.; Taylor, R.N.; Musci, T.J.; Rodgers, G.M.; Hubel, C.A.; McLaughlin, M.K. Preeclampsia: An endothelial cell disorder. Am. J. Obstet. Gynecol. 1989, 161, 1200–1204. [Google Scholar] [CrossRef] [PubMed]
  145. Hubel, C.A.; Roberts, J.M.; Taylor, R.N.; Musci, T.J.; Rogers, G.M.; McLaughlin, M.K. Lipid peroxidation in pregnancy: New perspectives on preeclampsia. Am. J. Obstet. Gynecol. 1989, 161, 1025–1034. [Google Scholar] [CrossRef]
  146. Many, A.; Hubel, C.A.; Fisher, S.J.; Roberts, J.M.; Zhou, Y. Invasive cytotrophoblasts manifest evidence of oxidative stress in preeclampsia. Am. J. Pathol. 2000, 156, 321–331. [Google Scholar] [CrossRef]
  147. Burton, G.J.; Yung, H.W.; Cindrova-Davies, T.; Charnock-Jones, D.S. Placental endoplasmic reticulum stress and oxidative stress in the pathophysiology of unexplained intrauterine growth restriction and early onset preeclampsia. Placenta 2009, 30 (Suppl. A), S43–S48. [Google Scholar] [CrossRef]
  148. Vangrieken, P.; Al-Nasiry, S.; Bast, A.; Leermakers, P.A.; Tulen, C.B.M.; Schiffers, P.M.H.; van Schooten, F.J.; Remels, A.H.V. Placental Mitochondrial Abnormalities in Preeclampsia. Reprod. Sci. 2021, 28, 2186–2199. [Google Scholar] [CrossRef] [PubMed]
  149. Harman, D. Free radical theory of aging. Mutat. Res. 1992, 275, 257–266. [Google Scholar] [CrossRef] [PubMed]
  150. Campagna, R.; Mazzanti, L.; Pompei, V.; Alia, S.; Vignini, A.; Emanuelli, M. The Multifaceted Role of Endothelial Sirt1 in Vascular Aging: An Update. Cells 2024, 13, 1469. [Google Scholar] [CrossRef]
  151. Hill, N.R.; Fatoba, S.T.; Oke, J.L.; Hirst, J.A.; O’Callaghan, C.A.; Lasserson, D.S.; Hobbs, F.D. Global Prevalence of Chronic Kidney Disease—A Systematic Review and Meta-Analysis. PLoS ONE 2016, 11, e0158765. [Google Scholar] [CrossRef]
  152. Sturmlechner, I.; Durik, M.; Sieben, C.J.; Baker, D.J.; van Deursen, J.M. Cellular senescence in renal ageing and disease. Nat. Rev. Nephrol. 2017, 13, 77–89. [Google Scholar] [CrossRef]
  153. Yamamoto, T.; Takabatake, Y.; Kimura, T.; Takahashi, A.; Namba, T.; Matsuda, J.; Minami, S.; Kaimori, J.Y.; Matsui, I.; Kitamura, H.; et al. Time-dependent dysregulation of autophagy: Implications in aging and mitochondrial homeostasis in the kidney proximal tubule. Autophagy 2016, 12, 801–813. [Google Scholar] [CrossRef]
  154. Martin-Maestro, P.; Gargini, R.; Perry, G.; Avila, J.; Garcia-Escudero, V. PARK2 enhancement is able to compensate mitophagy alterations found in sporadic Alzheimer’s disease. Hum. Mol. Genet. 2016, 25, 792–806. [Google Scholar] [CrossRef] [PubMed]
  155. Ravikumar, P.; Ye, J.; Zhang, J.; Pinch, S.N.; Hu, M.C.; Kuro-o, M.; Hsia, C.C.; Moe, O.W. alpha-Klotho protects against oxidative damage in pulmonary epithelia. Am. J. Physiol. Lung Cell Mol. Physiol. 2014, 307, L566–L575. [Google Scholar] [CrossRef]
  156. Kuro-o, M.; Matsumura, Y.; Aizawa, H.; Kawaguchi, H.; Suga, T.; Utsugi, T.; Ohyama, Y.; Kurabayashi, M.; Kaname, T.; Kume, E.; et al. Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature 1997, 390, 45–51. [Google Scholar] [CrossRef] [PubMed]
  157. Ito, S.; Kinoshita, S.; Shiraishi, N.; Nakagawa, S.; Sekine, S.; Fujimori, T.; Nabeshima, Y.I. Molecular cloning and expression analyses of mouse betaklotho, which encodes a novel Klotho family protein. Mech. Dev. 2000, 98, 115–119. [Google Scholar] [CrossRef]
  158. Ogawa, Y.; Kurosu, H.; Yamamoto, M.; Nandi, A.; Rosenblatt, K.P.; Goetz, R.; Eliseenkova, A.V.; Mohammadi, M.; Kuro-o, M. BetaKlotho is required for metabolic activity of fibroblast growth factor 21. Proc. Natl. Acad. Sci. USA 2007, 104, 7432–7437. [Google Scholar] [CrossRef] [PubMed]
  159. Bloch, L.; Sineshchekova, O.; Reichenbach, D.; Reiss, K.; Saftig, P.; Kuro-o, M.; Kaether, C. Klotho is a substrate for alpha-, beta- and gamma-secretase. FEBS Lett. 2009, 583, 3221–3224. [Google Scholar] [CrossRef] [PubMed]
  160. Imura, A.; Iwano, A.; Tohyama, O.; Tsuji, Y.; Nozaki, K.; Hashimoto, N.; Fujimori, T.; Nabeshima, Y. Secreted Klotho protein in sera and CSF: Implication for post-translational cleavage in release of Klotho protein from cell membrane. FEBS Lett. 2004, 565, 143–147. [Google Scholar] [CrossRef]
  161. Matsumura, Y.; Aizawa, H.; Shiraki-Iida, T.; Nagai, R.; Kuro-o, M.; Nabeshima, Y. Identification of the human klotho gene and its two transcripts encoding membrane and secreted klotho protein. Biochem. Biophys. Res. Commun. 1998, 242, 626–630. [Google Scholar] [CrossRef] [PubMed]
  162. Mencke, R.; Harms, G.; Moser, J.; van Meurs, M.; Diepstra, A.; Leuvenink, H.G.; Hillebrands, J.L. Human alternative Klotho mRNA is a nonsense-mediated mRNA decay target inefficiently spliced in renal disease. JCI Insight 2017, 2, e94375. [Google Scholar] [CrossRef] [PubMed]
  163. Kurosu, H.; Yamamoto, M.; Clark, J.D.; Pastor, J.V.; Nandi, A.; Gurnani, P.; McGuinness, O.P.; Chikuda, H.; Yamaguchi, M.; Kawaguchi, H.; et al. Suppression of aging in mice by the hormone Klotho. Science 2005, 309, 1829–1833. [Google Scholar] [CrossRef]
  164. Akimoto, T.; Yoshizawa, H.; Watanabe, Y.; Numata, A.; Yamazaki, T.; Takeshima, E.; Iwazu, K.; Komada, T.; Otani, N.; Morishita, Y.; et al. Characteristics of urinary and serum soluble Klotho protein in patients with different degrees of chronic kidney disease. BMC Nephrol. 2012, 13, 155. [Google Scholar] [CrossRef] [PubMed]
  165. Nagai, T.; Yamada, K.; Kim, H.C.; Kim, Y.S.; Noda, Y.; Imura, A.; Nabeshima, Y.; Nabeshima, T. Cognition impairment in the genetic model of aging klotho gene mutant mice: A role of oxidative stress. FASEB J. 2003, 17, 50–52. [Google Scholar] [CrossRef] [PubMed]
  166. Yamamoto, M.; Clark, J.D.; Pastor, J.V.; Gurnani, P.; Nandi, A.; Kurosu, H.; Miyoshi, M.; Ogawa, Y.; Castrillon, D.H.; Rosenblatt, K.P.; et al. Regulation of oxidative stress by the anti-aging hormone klotho. J. Biol. Chem. 2005, 280, 38029–38034. [Google Scholar] [CrossRef]
  167. Hu, M.C.; Shi, M.; Zhang, J.; Quinones, H.; Kuro-o, M.; Moe, O.W. Klotho deficiency is an early biomarker of renal ischemia-reperfusion injury and its replacement is protective. Kidney Int. 2010, 78, 1240–1251. [Google Scholar] [CrossRef] [PubMed]
  168. Sabren, S.; Hagar, T.; Khateeb, N.; Evgeny, F.; Yara, F.N.; Perlitz, Y.; Farid, N. Placental and serum levels of human alpha-Klotho in preeclampsia & intra-uterine growth retardation: A potential sensitive biomarker? Pregnancy Hypertens. 2024, 36, 101115. [Google Scholar] [CrossRef]
  169. Hsia, C.C.W.; Ravikumar, P.; Ye, J. Acute lung injury complicating acute kidney injury: A model of endogenous alphaKlotho deficiency and distant organ dysfunction. Bone 2017, 100, 100–109. [Google Scholar] [CrossRef] [PubMed]
  170. Buendia-Roldan, I.; Machuca, N.; Mejia, M.; Maldonado, M.; Pardo, A.; Selman, M. Lower levels of alpha-Klotho in serum are associated with decreased lung function in individuals with interstitial lung abnormalities. Sci. Rep. 2019, 9, 10801. [Google Scholar] [CrossRef]
  171. Pako, J.; Bikov, A.; Barta, I.; Matsueda, H.; Puskas, R.; Galffy, G.; Kerpel-Fronius, A.; Antus, B.; Horvath, I. Assessment of the circulating klotho protein in lung cancer patients. Pathol. Oncol. Res. 2020, 26, 233–238. [Google Scholar] [CrossRef]
  172. Keller, J.N.; Schmitt, F.A.; Scheff, S.W.; Ding, Q.; Chen, Q.; Butterfield, D.A.; Markesbery, W.R. Evidence of increased oxidative damage in subjects with mild cognitive impairment. Neurology 2005, 64, 1152–1156. [Google Scholar] [CrossRef] [PubMed]
  173. Barnham, K.J.; Masters, C.L.; Bush, A.I. Neurodegenerative diseases and oxidative stress. Nat. Rev. Drug Discov. 2004, 3, 205–214. [Google Scholar] [CrossRef] [PubMed]
  174. Semba, R.D.; Moghekar, A.R.; Hu, J.; Sun, K.; Turner, R.; Ferrucci, L.; O’Brien, R. Klotho in the cerebrospinal fluid of adults with and without Alzheimer’s disease. Neurosci. Lett. 2014, 558, 37–40. [Google Scholar] [CrossRef] [PubMed]
  175. Shardell, M.; Semba, R.D.; Rosano, C.; Kalyani, R.R.; Bandinelli, S.; Chia, C.W.; Ferrucci, L. Plasma Klotho and Cognitive Decline in Older Adults: Findings From the InCHIANTI Study. J. Gerontol. A Biol. Sci. Med. Sci. 2016, 71, 677–682. [Google Scholar] [CrossRef]
  176. Lim, K.; Groen, A.; Molostvov, G.; Lu, T.; Lilley, K.S.; Snead, D.; James, S.; Wilkinson, I.B.; Ting, S.; Hsiao, L.L.; et al. alpha-Klotho Expression in Human Tissues. J. Clin. Endocrinol. Metab. 2015, 100, E1308–E1318. [Google Scholar] [CrossRef]
  177. Rao, Z.; Landry, T.; Li, P.; Bunner, W.; Laing, B.T.; Yuan, Y.; Huang, H. Administration of alpha klotho reduces liver and adipose lipid accumulation in obese mice. Heliyon 2019, 5, e01494. [Google Scholar] [CrossRef]
  178. Szeto, A.; Cecati, M.; Ahmed, R.; McCabe, P.M.; Mendez, A.J. Oxytocin reduces adipose tissue inflammation in obese mice. Lipids Health Dis. 2020, 19, 188. [Google Scholar] [CrossRef] [PubMed]
  179. Mateuszuk, L.; Campagna, R.; Kutryb-Zajac, B.; Kus, K.; Slominska, E.M.; Smolenski, R.T.; Chlopicki, S. Reversal of endothelial dysfunction by nicotinamide mononucleotide via extracellular conversion to nicotinamide riboside. Biochem. Pharmacol. 2020, 178, 114019. [Google Scholar] [CrossRef]
  180. Campagna, R.; Mateuszuk, L.; Wojnar-Lason, K.; Kaczara, P.; Tworzydlo, A.; Kij, A.; Bujok, R.; Mlynarski, J.; Wang, Y.; Sartini, D.; et al. Nicotinamide N-methyltransferase in endothelium protects against oxidant stress-induced endothelial injury. Biochim. Biophys. Acta Mol. Cell Res. 2021, 1868, 119082. [Google Scholar] [CrossRef] [PubMed]
  181. Tousoulis, D.; Kampoli, A.M.; Tentolouris, C.; Papageorgiou, N.; Stefanadis, C. The role of nitric oxide on endothelial function. Curr. Vasc. Pharmacol. 2012, 10, 4–18. [Google Scholar] [CrossRef] [PubMed]
  182. Saito, Y.; Yamagishi, T.; Nakamura, T.; Ohyama, Y.; Aizawa, H.; Suga, T.; Matsumura, Y.; Masuda, H.; Kurabayashi, M.; Kuro-o, M.; et al. Klotho protein protects against endothelial dysfunction. Biochem. Biophys. Res. Commun. 1998, 248, 324–329. [Google Scholar] [CrossRef]
  183. Nagai, R.; Saito, Y.; Ohyama, Y.; Aizawa, H.; Suga, T.; Nakamura, T.; Kurabayashi, M.; Kuroo, M. Endothelial dysfunction in the klotho mouse and downregulation of klotho gene expression in various animal models of vascular and metabolic diseases. Cell Mol. Life Sci. 2000, 57, 738–746. [Google Scholar] [CrossRef] [PubMed]
  184. Chung, C.P.; Chang, Y.C.; Ding, Y.; Lim, K.; Liu, Q.; Zhu, L.; Zhang, W.; Lu, T.S.; Molostvov, G.; Zehnder, D.; et al. alpha-Klotho expression determines nitric oxide synthesis in response to FGF-23 in human aortic endothelial cells. PLoS ONE 2017, 12, e0176817. [Google Scholar] [CrossRef] [PubMed]
  185. Liu, Y.; Chen, M. Emerging role of alpha-Klotho in energy metabolism and cardiometabolic diseases. Diabetes Metab. Syndr. 2023, 17, 102854. [Google Scholar] [CrossRef]
  186. Begum, M.K.; Konja, D.; Singh, S.; Chlopicki, S.; Wang, Y. Endothelial SIRT1 as a Target for the Prevention of Arterial Aging: Promises and Challenges. J. Cardiovasc. Pharmacol. 2021, 78, S63–S77. [Google Scholar] [CrossRef] [PubMed]
  187. Kida, Y.; Goligorsky, M.S. Sirtuins, Cell Senescence, and Vascular Aging. Can. J. Cardiol. 2016, 32, 634–641. [Google Scholar] [CrossRef] [PubMed]
  188. Gao, D.; Zuo, Z.; Tian, J.; Ali, Q.; Lin, Y.; Lei, H.; Sun, Z. Activation of SIRT1 Attenuates Klotho Deficiency-Induced Arterial Stiffness and Hypertension by Enhancing AMP-Activated Protein Kinase Activity. Hypertension 2016, 68, 1191–1199. [Google Scholar] [CrossRef]
  189. Fujitsuka, N.; Asakawa, A.; Morinaga, A.; Amitani, M.S.; Amitani, H.; Katsuura, G.; Sawada, Y.; Sudo, Y.; Uezono, Y.; Mochiki, E.; et al. Increased ghrelin signaling prolongs survival in mouse models of human aging through activation of sirtuin1. Mol. Psychiatry 2016, 21, 1613–1623. [Google Scholar] [CrossRef]
  190. Li, C.; Jiang, S.; Wang, H.; Wang, Y.; Han, Y.; Jiang, J. Berberine exerts protective effects on cardiac senescence by regulating the Klotho/SIRT1 signaling pathway. Biomed. Pharmacother. 2022, 151, 113097. [Google Scholar] [CrossRef]
  191. Han, J.; Lu, Z.; Qi, Y.; Liu, T.; Li, Y.; Han, H.; Zhao, C.; Ma, X. Melatonin Attenuates PFOS-Induced Reproductive Toxicity of Pregnant Mice due to Placental Damage via Antioxidant, Anti-Aging and Anti-Inflammatory Pathways. Birth Defects Res. 2024, 116, e2423. [Google Scholar] [CrossRef] [PubMed]
  192. Lee, S.; Jung, M.H.; Song, K.; Jin, J.X.; Taweechaipaisankul, A.; Kim, G.A.; Oh, H.J.; Koo, O.J.; Park, S.C.; Lee, B.C. Failure to maintain full-term pregnancies in pig carrying klotho monoallelic knockout fetuses. BMC Biotechnol. 2021, 21, 1. [Google Scholar] [CrossRef]
  193. Iurciuc, S.; Cimpean, A.M.; Mitu, F.; Heredea, R.; Iurciuc, M. Vascular aging and subclinical atherosclerosis: Why such a “never ending” and challenging story in cardiology? Clin. Interv. Aging 2017, 12, 1339–1345. [Google Scholar] [CrossRef] [PubMed]
  194. Ohata, Y.; Arahori, H.; Namba, N.; Kitaoka, T.; Hirai, H.; Wada, K.; Nakayama, M.; Michigami, T.; Imura, A.; Nabeshima, Y.; et al. Circulating levels of soluble alpha-Klotho are markedly elevated in human umbilical cord blood. J. Clin. Endocrinol. Metab. 2011, 96, E943–E947. [Google Scholar] [CrossRef]
  195. Shimada, T.; Takeshita, Y.; Murohara, T.; Sasaki, K.; Egami, K.; Shintani, S.; Katsuda, Y.; Ikeda, H.; Nabeshima, Y.; Imaizumi, T. Angiogenesis and vasculogenesis are impaired in the precocious-aging klotho mouse. Circulation 2004, 110, 1148–1155. [Google Scholar] [CrossRef] [PubMed]
  196. Razzaque, M.S. The role of Klotho in energy metabolism. Nat. Rev. Endocrinol. 2012, 8, 579–587. [Google Scholar] [CrossRef]
  197. Woudenberg-Vrenken, T.E.; van der Eerden, B.C.; van der Kemp, A.W.; van Leeuwen, J.P.; Bindels, R.J.; Hoenderop, J.G. Characterization of vitamin D-deficient klotho(-/-) mice: Do increased levels of serum 1,25(OH)2D3 cause disturbed calcium and phosphate homeostasis in klotho(-/-) mice? Nephrol. Dial. Transplant. 2012, 27, 4061–4068. [Google Scholar] [CrossRef] [PubMed]
  198. Nakatani, T.; Sarraj, B.; Ohnishi, M.; Densmore, M.J.; Taguchi, T.; Goetz, R.; Mohammadi, M.; Lanske, B.; Razzaque, M.S. In vivo genetic evidence for klotho-dependent, fibroblast growth factor 23 (Fgf23) -mediated regulation of systemic phosphate homeostasis. FASEB J. 2009, 23, 433–441. [Google Scholar] [CrossRef] [PubMed]
  199. Godang, K.; Froslie, K.F.; Henriksen, T.; Isaksen, G.A.; Voldner, N.; Lekva, T.; Ueland, T.; Bollerslev, J. Umbilical cord levels of sclerostin, placental weight, and birth weight are predictors of total bone mineral content in neonates. Eur. J. Endocrinol. 2013, 168, 371–378. [Google Scholar] [CrossRef]
  200. Ma, R.; Gu, Y.; Groome, L.J.; Wang, Y. ADAM17 regulates TNFalpha production by placental trophoblasts. Placenta 2011, 32, 975–980. [Google Scholar] [CrossRef]
  201. Ohata, Y.; Yamazaki, M.; Kawai, M.; Tsugawa, N.; Tachikawa, K.; Koinuma, T.; Miyagawa, K.; Kimoto, A.; Nakayama, M.; Namba, N.; et al. Elevated fibroblast growth factor 23 exerts its effects on placenta and regulates vitamin D metabolism in pregnancy of Hyp mice. J. Bone Miner. Res. 2014, 29, 1627–1638. [Google Scholar] [CrossRef]
  202. Yang, Y.; Wang, Y.; Zeng, X.; Ma, X.J.; Zhao, Y.; Qiao, J.; Cao, B.; Li, Y.X.; Ji, L.; Wang, Y.L. Self-control of HGF regulation on human trophoblast cell invasion via enhancing c-Met receptor shedding by ADAM10 and ADAM17. J. Clin. Endocrinol. Metab. 2012, 97, E1390–E1401. [Google Scholar] [CrossRef] [PubMed]
  203. Fantone, S.; Ermini, L.; Piani, F.; Di Simone, N.; Barbaro, G.; Giannubilo, S.R.; Gesuita, R.; Tossetta, G.; Marzioni, D. Downregulation of argininosuccinate synthase 1 (ASS1) is associated with hypoxia in placental development. Hum. Cell 2023, 36, 1190–1198. [Google Scholar] [CrossRef] [PubMed]
  204. Tossetta, G.; Fantone, S.; Busilacchi, E.M.; Di Simone, N.; Giannubilo, S.R.; Scambia, G.; Giordano, A.; Marzioni, D. Modulation of matrix metalloproteases by ciliary neurotrophic factor in human placental development. Cell Tissue Res. 2022, 390, 113–129. [Google Scholar] [CrossRef] [PubMed]
  205. Tossetta, G.; Avellini, C.; Licini, C.; Giannubilo, S.R.; Castellucci, M.; Marzioni, D. High temperature requirement A1 and fibronectin: Two possible players in placental tissue remodelling. Eur. J. Histochem. 2016, 60, 2724. [Google Scholar] [CrossRef]
  206. Miranda, J.; Romero, R.; Korzeniewski, S.J.; Schwartz, A.G.; Chaemsaithong, P.; Stampalija, T.; Yeo, L.; Dong, Z.; Hassan, S.S.; Chrousos, G.P.; et al. The anti-aging factor alpha-klotho during human pregnancy and its expression in pregnancies complicated by small-for-gestational-age neonates and/or preeclampsia. J. Matern. Fetal Neonatal Med. 2014, 27, 449–457. [Google Scholar] [CrossRef]
  207. Shao, W.J.; Wang, D.X.; Wan, Q.Y.; Zhang, M.M.; Chen, M.M.; Song, W.W. Expression of mRNA and protein of Klotho gene in placental tissue of macrosomia and its relationship with birth weight of neonates. Zhonghua Fu Chan Ke Za Zhi 2016, 51, 420–423. [Google Scholar] [CrossRef] [PubMed]
  208. Fan, C.; Wang, Y.; Wang, J.; Lei, D.; Sun, Y.; Lei, S.; Hu, M.; Tian, Y.; Li, R.; Wang, S. Clinic significance of markedly decreased alpha-klothoin women with preeclampsia. Am. J. Transl. Res. 2016, 8, 1998–2010. [Google Scholar] [PubMed]
  209. Wang, J.; Lin, Y.; Lv, F.; Hou, H.; Chu, K.; Jiang, B.; Liu, J.; Liu, S.; Hou, B. Effects of Klotho polymorphisms on Preeclampsia risk in a case-control study. Pregnancy Hypertens. 2018, 13, 95–99. [Google Scholar] [CrossRef] [PubMed]
  210. Fogarty, N.M.; Ferguson-Smith, A.C.; Burton, G.J. Syncytial knots (Tenney-Parker changes) in the human placenta: Evidence of loss of transcriptional activity and oxidative damage. Am. J. Pathol. 2013, 183, 144–152. [Google Scholar] [CrossRef]
  211. Goldman-Wohl, D.; Yagel, S. United we stand not dividing: The syncytiotrophoblast and cell senescence. Placenta 2014, 35, 341–344. [Google Scholar] [CrossRef] [PubMed]
  212. de Oliveira, R.M. Klotho RNAi induces premature senescence of human cells via a p53/p21 dependent pathway. FEBS Lett. 2006, 580, 5753–5758. [Google Scholar] [CrossRef] [PubMed]
  213. Loichinger, M.H.; Towner, D.; Thompson, K.S.; Ahn, H.J.; Bryant-Greenwood, G.D. Systemic and placental alpha-klotho: Effects of preeclampsia in the last trimester of gestation. Placenta 2016, 41, 53–61. [Google Scholar] [CrossRef] [PubMed]
  214. Buendia, I.; Michalska, P.; Navarro, E.; Gameiro, I.; Egea, J.; Leon, R. Nrf2-ARE pathway: An emerging target against oxidative stress and neuroinflammation in neurodegenerative diseases. Pharmacol. Ther. 2016, 157, 84–104. [Google Scholar] [CrossRef] [PubMed]
  215. Chen, K.; Wang, S.; Sun, Q.W.; Zhang, B.; Ullah, M.; Sun, Z. Klotho Deficiency Causes Heart Aging via Impairing the Nrf2-GR Pathway. Circ. Res. 2021, 128, 492–507. [Google Scholar] [CrossRef]
  216. Xing, L.; Guo, H.; Meng, S.; Zhu, B.; Fang, J.; Huang, J.; Chen, J.; Wang, Y.; Wang, L.; Yao, X.; et al. Klotho ameliorates diabetic nephropathy by activating Nrf2 signaling pathway in podocytes. Biochem. Biophys. Res. Commun. 2021, 534, 450–456. [Google Scholar] [CrossRef] [PubMed]
  217. Xu, B.; Cheng, F.; Xue, X. Klotho-mediated activation of the anti-oxidant Nrf2/ARE signal pathway affects cell apoptosis, senescence and mobility in hypoxic human trophoblasts: Involvement of Klotho in the pathogenesis of preeclampsia. Cell Div. 2024, 19, 13. [Google Scholar] [CrossRef] [PubMed]
  218. Barbieri, M.; Bonafe, M.; Franceschi, C.; Paolisso, G. Insulin/IGF-I-signaling pathway: An evolutionarily conserved mechanism of longevity from yeast to humans. Am. J. Physiol. Endocrinol. Metab. 2003, 285, E1064–E1071. [Google Scholar] [CrossRef] [PubMed]
  219. Salmon, W.D., Jr.; Daughaday, W.H. A hormonally controlled serum factor which stimulates sulfate incorporation by cartilage in vitro. J. Lab. Clin. Med. 1957, 49, 825–836. [Google Scholar]
  220. Tricoli, J.V.; Rall, L.B.; Scott, J.; Bell, G.I.; Shows, T.B. Localization of insulin-like growth factor genes to human chromosomes 11 and 12. Nature 1984, 310, 784–786. [Google Scholar] [CrossRef]
  221. Adamo, M.L.; Ben-Hur, H.; Roberts, C.T., Jr.; LeRoith, D. Regulation of start site usage in the leader exons of the rat insulin-like growth factor-I gene by development, fasting, and diabetes. Mol. Endocrinol. 1991, 5, 1677–1686. [Google Scholar] [CrossRef] [PubMed]
  222. Werner, H.; Adamo, M.; Roberts, C.T., Jr.; LeRoith, D. Molecular and cellular aspects of insulin-like growth factor action. Vitam. Horm. 1994, 48, 1–58. [Google Scholar] [CrossRef] [PubMed]
  223. Sussenbach, J.S. The gene structure of the insulin-like growth factor family. Prog. Growth Factor. Res. 1989, 1, 33–48. [Google Scholar] [CrossRef]
  224. Delafontaine, P.; Song, Y.H.; Li, Y. Expression, regulation, and function of IGF-1, IGF-1R, and IGF-1 binding proteins in blood vessels. Arterioscler. Thromb. Vasc. Biol. 2004, 24, 435–444. [Google Scholar] [CrossRef] [PubMed]
  225. Delafontaine, P. Insulin-like growth factor I and its binding proteins in the cardiovascular system. Cardiovasc. Res. 1995, 30, 825–834. [Google Scholar] [CrossRef]
  226. Le Roith, D. Seminars in medicine of the Beth Israel Deaconess Medical Center. Insulin-like growth factors. N. Engl. J. Med. 1997, 336, 633–640. [Google Scholar] [CrossRef] [PubMed]
  227. Peterson, J.E.; Kulik, G.; Jelinek, T.; Reuter, C.W.; Shannon, J.A.; Weber, M.J. Src phosphorylates the insulin-like growth factor type I receptor on the autophosphorylation sites. Requirement for transformation by src. J. Biol. Chem. 1996, 271, 31562–31571. [Google Scholar] [CrossRef] [PubMed]
  228. Tsuruzoe, K.; Emkey, R.; Kriauciunas, K.M.; Ueki, K.; Kahn, C.R. Insulin receptor substrate 3 (IRS-3) and IRS-4 impair IRS-1- and IRS-2-mediated signaling. Mol. Cell Biol. 2001, 21, 26–38. [Google Scholar] [CrossRef] [PubMed]
  229. Saltiel, A.R.; Kahn, C.R. Insulin signalling and the regulation of glucose and lipid metabolism. Nature 2001, 414, 799–806. [Google Scholar] [CrossRef] [PubMed]
  230. Bhullar, K.S.; Lagaron, N.O.; McGowan, E.M.; Parmar, I.; Jha, A.; Hubbard, B.P.; Rupasinghe, H.P.V. Kinase-targeted cancer therapies: Progress, challenges and future directions. Mol. Cancer 2018, 17, 48. [Google Scholar] [CrossRef]
  231. Szymonowicz, K.; Oeck, S.; Malewicz, N.M.; Jendrossek, V. New Insights into Protein Kinase B/Akt Signaling: Role of Localized Akt Activation and Compartment-Specific Target Proteins for the Cellular Radiation Response. Cancers 2018, 10, 78. [Google Scholar] [CrossRef] [PubMed]
  232. Zhang, X.; Tang, N.; Hadden, T.J.; Rishi, A.K. Akt, FoxO and regulation of apoptosis. Biochim. Biophys. Acta 2011, 1813, 1978–1986. [Google Scholar] [CrossRef] [PubMed]
  233. Kaufmann, E.; Knochel, W. Five years on the wings of fork head. Mech. Dev. 1996, 57, 3–20. [Google Scholar] [CrossRef] [PubMed]
  234. Zecic, A.; Braeckman, B.P. DAF-16/FoxO in Caenorhabditis elegans and Its Role in Metabolic Remodeling. Cells 2020, 9, 109. [Google Scholar] [CrossRef]
  235. Tuteja, G.; Kaestner, K.H. SnapShot: Forkhead transcription factors I. Cell 2007, 130, 1160. [Google Scholar] [CrossRef] [PubMed]
  236. Lappas, M.; Lim, R.; Riley, C.; Menon, R.; Permezel, M. Expression and localisation of FoxO3 and FoxO4 in human placenta and fetal membranes. Placenta 2010, 31, 1043–1050. [Google Scholar] [CrossRef]
  237. Xu, Y.; Jin, B.; Sun, L.; Yang, H.; Cao, X.; Zhang, G. The expression of FoxO1 in placenta and omental adipose tissue of gestational diabetes mellitus. Exp. Clin. Endocrinol. Diabetes 2014, 122, 287–294. [Google Scholar] [CrossRef] [PubMed]
  238. Kato, M.; Yuan, H.; Xu, Z.G.; Lanting, L.; Li, S.L.; Wang, M.; Hu, M.C.; Reddy, M.A.; Natarajan, R. Role of the Akt/FoxO3a pathway in TGF-beta1-mediated mesangial cell dysfunction: A novel mechanism related to diabetic kidney disease. J. Am. Soc. Nephrol. 2006, 17, 3325–3335. [Google Scholar] [CrossRef] [PubMed]
  239. Mao, Z.; Liu, L.; Zhang, R.; Li, X. Lithium reduces FoxO3a transcriptional activity by decreasing its intracellular content. Biol. Psychiatry 2007, 62, 1423–1430. [Google Scholar] [CrossRef] [PubMed]
  240. Tremblay, M.L.; Giguere, V. Phosphatases at the heart of FoxO metabolic control. Cell Metab. 2008, 7, 101–103. [Google Scholar] [CrossRef] [PubMed]
  241. Ji, F.; Chen, R.; Liu, B.; Zhang, X.; Han, J.; Wang, H.; Shen, G.; Tao, J. BAFF induces spleen CD4+ T cell proliferation by down-regulating phosphorylation of FOXO3A and activates cyclin D2 and D3 expression. Biochem. Biophys. Res. Commun. 2012, 425, 854–858. [Google Scholar] [CrossRef] [PubMed]
  242. Shenker, J.J.; Sengupta, S.M.; Joober, R.; Malla, A.; Chakravarty, M.M.; Lepage, M. Bipolar disorder risk gene FOXO6 modulates negative symptoms in schizophrenia: A neuroimaging genetics study. J. Psychiatry Neurosci. 2017, 42, 172–180. [Google Scholar] [CrossRef]
  243. Lim, S.W.; Jin, L.; Luo, K.; Jin, J.; Shin, Y.J.; Hong, S.Y.; Yang, C.W. Klotho enhances FoxO3-mediated manganese superoxide dismutase expression by negatively regulating PI3K/AKT pathway during tacrolimus-induced oxidative stress. Cell Death Dis. 2017, 8, e2972. [Google Scholar] [CrossRef] [PubMed]
  244. Sakaguchi, M. The role of insulin signaling with FOXO and FOXK transcription factors. Endocr. J. 2024, 71, 939–944. [Google Scholar] [CrossRef] [PubMed]
  245. Orellana, A.M.; Mazucanti, C.H.; Dos Anjos, L.P.; de Sa Lima, L.; Kawamoto, E.M.; Scavone, C. Klotho increases antioxidant defenses in astrocytes and ubiquitin-proteasome activity in neurons. Sci. Rep. 2023, 13, 15080. [Google Scholar] [CrossRef] [PubMed]
  246. Hu, M.C.; Shi, M.; Cho, H.J.; Zhang, J.; Pavlenco, A.; Liu, S.; Sidhu, S.; Huang, L.J.; Moe, O.W. The erythropoietin receptor is a downstream effector of Klotho-induced cytoprotection. Kidney Int. 2013, 84, 468–481. [Google Scholar] [CrossRef] [PubMed]
  247. Zhou, H.J.; Zeng, C.Y.; Yang, T.T.; Long, F.Y.; Kuang, X.; Du, J.R. Lentivirus-mediated klotho up-regulation improves aging-related memory deficits and oxidative stress in senescence-accelerated mouse prone-8 mice. Life Sci. 2018, 200, 56–62. [Google Scholar] [CrossRef] [PubMed]
  248. Leduc-Gaudet, J.P.; Franco-Romero, A.; Cefis, M.; Moamer, A.; Broering, F.E.; Milan, G.; Sartori, R.; Chaffer, T.J.; Dulac, M.; Marcangeli, V.; et al. MYTHO is a novel regulator of skeletal muscle autophagy and integrity. Nat. Commun. 2023, 14, 1199. [Google Scholar] [CrossRef] [PubMed]
  249. Cox, L.S.; Redman, C. The role of cellular senescence in ageing of the placenta. Placenta 2017, 52, 139–145. [Google Scholar] [CrossRef]
  250. Pollack, A.Z.; Rivers, K.; Ahrens, K.A. Parity associated with telomere length among US reproductive age women. Hum. Reprod. 2018, 33, 736–744. [Google Scholar] [CrossRef]
  251. Iqbal, S.; Lockett, G.A.; Holloway, J.W.; Arshad, S.H.; Zhang, H.; Kaushal, A.; Tetali, S.R.; Mukherjee, N.; Karmaus, W.J.J. Changes in DNA Methylation from Age 18 to Pregnancy in Type 1, 2, and 17 T Helper and Regulatory T-Cells Pathway Genes. Int. J. Mol. Sci. 2018, 19, 477. [Google Scholar] [CrossRef] [PubMed]
  252. Giller, A.; Andrawus, M.; Gutman, D.; Atzmon, G. Pregnancy as a model for aging. Ageing Res. Rev. 2020, 62, 101093. [Google Scholar] [CrossRef]
  253. Mensa, E.; Guescini, M.; Giuliani, A.; Bacalini, M.G.; Ramini, D.; Corleone, G.; Ferracin, M.; Fulgenzi, G.; Graciotti, L.; Prattichizzo, F.; et al. Small extracellular vesicles deliver miR-21 and miR-217 as pro-senescence effectors to endothelial cells. J. Extracell. Vesicles 2020, 9, 1725285. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Soluble form of Klotho (sKlotho) and antioxidant mechanisms. Once the membrane-anchored secretases AD-AM10/17 cleaves the membrane form of Klotho (mKlotho), the extracellular domain is released into the extracellular space. The extracellular domain represents the soluble form of Klotho (sKlotho) which, via peripheral circulation, reaches the cell target and inhibits the insulin/IGF-1/PI3K/Akt/FoxO pathway. Therefore, the expression of genes related to antioxidant defense (such as superoxide dismutase (SOD2) and catalase (CAT)) is promoted and the oxidative stress decreases. Similarly, FoxO proteins induce the transcription of the Mytho gene, a regulator of autophagy and skeletal muscle integrity.
Figure 1. Soluble form of Klotho (sKlotho) and antioxidant mechanisms. Once the membrane-anchored secretases AD-AM10/17 cleaves the membrane form of Klotho (mKlotho), the extracellular domain is released into the extracellular space. The extracellular domain represents the soluble form of Klotho (sKlotho) which, via peripheral circulation, reaches the cell target and inhibits the insulin/IGF-1/PI3K/Akt/FoxO pathway. Therefore, the expression of genes related to antioxidant defense (such as superoxide dismutase (SOD2) and catalase (CAT)) is promoted and the oxidative stress decreases. Similarly, FoxO proteins induce the transcription of the Mytho gene, a regulator of autophagy and skeletal muscle integrity.
Ijms 26 00902 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Cecati, M.; Fumarola, S.; Vaiasicca, S.; Cianfruglia, L.; Vignini, A.; Giannubilo, S.R.; Emanuelli, M.; Ciavattini, A. Preeclampsia as a Study Model for Aging: The Klotho Gene Paradigm. Int. J. Mol. Sci. 2025, 26, 902. https://doi.org/10.3390/ijms26030902

AMA Style

Cecati M, Fumarola S, Vaiasicca S, Cianfruglia L, Vignini A, Giannubilo SR, Emanuelli M, Ciavattini A. Preeclampsia as a Study Model for Aging: The Klotho Gene Paradigm. International Journal of Molecular Sciences. 2025; 26(3):902. https://doi.org/10.3390/ijms26030902

Chicago/Turabian Style

Cecati, Monia, Stefania Fumarola, Salvatore Vaiasicca, Laura Cianfruglia, Arianna Vignini, Stefano Raffaele Giannubilo, Monica Emanuelli, and Andrea Ciavattini. 2025. "Preeclampsia as a Study Model for Aging: The Klotho Gene Paradigm" International Journal of Molecular Sciences 26, no. 3: 902. https://doi.org/10.3390/ijms26030902

APA Style

Cecati, M., Fumarola, S., Vaiasicca, S., Cianfruglia, L., Vignini, A., Giannubilo, S. R., Emanuelli, M., & Ciavattini, A. (2025). Preeclampsia as a Study Model for Aging: The Klotho Gene Paradigm. International Journal of Molecular Sciences, 26(3), 902. https://doi.org/10.3390/ijms26030902

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop