Next Article in Journal
Updates in Diagnostic Imaging for Infectious Keratitis: A Review
Previous Article in Journal
Relationship between Abdominal Volume Index and Body Adiposity Index and Scales of Insulin Resistance and Metabolic Syndrome
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Klotho in Cancer: Potential Diagnostic and Prognostic Applications

by
Jucileide Mota
1,
Alice Marques Moreira Lima
2,
Jhessica I. S. Gomes
1,
Marcelo Souza de Andrade
1,
Haissa O. Brito
1,3,
Melaine M. A. Lawall Silva
3,
Ana I. Faustino-Rocha
4,5,*,
Paula A. Oliveira
4,5,
Fernanda F. Lopes
1 and
Rui M. Gil da Costa
1,4,5,6,7,8,9
1
Post-Graduate Programme in Adult Health (PPGSAD), Federal University of Maranhão, São Luís 65085-580, Brazil
2
Health Sciences Center, State University of the Tocantins Region of Maranhão (UEMASUL), Imperatriz 6591-480, Brazil
3
Morphology Department, Federal University of Maranhão, São Luís 65085-580, Brazil
4
Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
5
Inov4Agro—Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
6
Laboratory for Process Engineering, Environment, Biotechnology and Energy (LEPABE), Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal
7
Associate Laboratory in Chemical Engineering, Faculty of Engineering (ALiCE), University of Porto, 4200-465 Porto, Portugal
8
Molecular Oncology and Viral Pathology Group, Portuguese Oncology Institute of Porto (IPO Porto), 4200-072 Porto, Portugal
9
Health Research Network, Research Center of Portuguese Oncology Institute of Porto (CIIPOP/RISE@CIIPOP), 4200-072 Porto, Portugal
*
Author to whom correspondence should be addressed.
Diagnostics 2023, 13(21), 3357; https://doi.org/10.3390/diagnostics13213357
Submission received: 26 September 2023 / Revised: 25 October 2023 / Accepted: 26 October 2023 / Published: 31 October 2023

Abstract

:
Klotho proteins, αKlotho, βKlotho, and γKlotho, exert tumor-suppressive activities via the fibroblast growth factor receptors and multiple cell-signaling pathways. There is a growing interest in Klotho proteins as potential diagnostic and prognostic biomarkers for multiple diseases. However, recent advances regarding their roles and potential applications in cancer remain disperse and require an integrated analysis. The present review analyzed research articles published between 2012 and 2022 in the Cochrane and Scopus scientific databases to study the role of Klotho in cancer and their potential as tools for diagnosing specific cancer types, predicting tumor aggressiveness and prognosis. Twenty-six articles were selected, dealing with acute myeloid leukemia and with bladder, breast, colorectal, esophageal, gastric, hepatocellular, ovarian, pancreatic, prostatic, pulmonary, renal, and thyroid cancers. αKlotho was consistently associated with improved prognosis and may be useful in estimating patient survival. A single study reported the use of soluble αKlotho levels in blood serum as a tool to aid the diagnosis of esophageal cancer. γKlotho was associated with increased aggressiveness of bladder, breast, and prostate cancer, and βKlotho showed mixed results. Further clinical development of Klotho-based assays will require careful identification of specific tumor subtypes where Klotho proteins may be most valuable as diagnostic or prognostic tools.

1. Introduction

The Klotho proteins, alpha(α)Klotho [1,2] and beta(β)Klotho [3], are encoded by the KLA and KLB genes located in chromosomes 4 and 13, respectively. αKlotho was originally identified in mice and elicited great interest due to its anti-aging properties [1]. It is expressed in a variety of tissues and is in the cell membrane as a type I single-pass 135 kDa protein containing an N-terminal sequence, two extracellular domains (designated KL1 and KL2) with glycosidase activity, a transmembrane helix, and an intracellular domain consisting of only 10 amino acids [2].
The αKlotho protein is also present in blood as a secreted protein generated by alternative mRNA splicing containing the KL1 domain only [1] and as a soluble protein that may contain KL1 alone or both the KL1 and Kl2 extracellular domains [4]. Cleavage of the αKlotho extracellular domains is mediated by disintegrin and metalloproteinase domain-containing (ADAM) proteins ADAM10 and ADAM17 [4]. The βKlotho protein shares structural similarities with αKlotho and is also located in the cell’s plasma membrane [3,5], and soluble βKlotho has also been reported [6]. Another membrane-bound glycosidase-like protein, designated Klotho-lactase phlorizin hydrolase, was first identified in mice and is encoded by the LCTL gene on chromosome 15 in humans [7]. The functions of this protein, also referred to as γKlotho, are less clear than those of αKlotho and βKlotho.
αKlotho binds to FGR receptors, acting as a co-receptor for FGF23 and playing a key role in the renal regulation of phosphate levels [8,9]. βKlotho acts as a co-receptor for fibroblast growth factors 19 and 21 (FGF19 and FGF21) by forming binary complexes with FGFR4 and FGFR1c, respectively [10,11,12]. The binding of βKlotho with FGFR1c in adipose tissue or FGFR4 in the liver and with endocrine ligands FGF21 and FGF19 triggers multiple intracellular responses, as previously reviewed [5]. Canonically, the binding of FGF21 to the βKlotho-FGFR1c complex activates ERK1/2 downstream signaling and regulates the synthesis of biliary acids in hepatocytes, while FGF19 binds to βKlotho-FGFR4 complexes to downregulate Cyp17a1, also regulating hepatic bile production [11,12,13,14].
Loss of αKlotho has been consistently linked with chronic kidney disease and phosphate metabolism dysfunction [15,16]. αKlotho downregulation was also associated with pleiotropic effects involved in aging [1,5] and is proposed to act as a tumor suppressor, as recently reviewed [17]. Interestingly, βKlotho has been associated with both tumorigenic and tumor-suppressive effects in different types of cancer, suggesting a more complex scenario with multiple context-specific activities [18,19,20]. γKlotho expression has also been studied in multiple types of cancer [21,22]. In cancer, Klotho proteins have been shown to interact with multiple cellular signaling pathways, enhancing or blocking carcinogenesis, as previously reviewed [17,23]. As well as interacting with FGF to activate FGFR, αKlotho (Figure 1) was initially found to downregulate signaling via insulin-like growth factor 1 receptor (IGF-1R), and this may contribute to its effects against some types of cancer [24,25]. βKlotho enhances pro-tumorigenic functions of FGFR in multiple types of cancer [26,27]. The phosphatidylinositol-3-kinase (PI3K) pathway is triggered by multiple membrane-bound receptors and mediates cell proliferation, growth, and survival and is also inhibited by αKlotho [28]. The WNT-β-catenin pathway is activated in multiple cancers where it modulates cell differentiation, survival, and mobility [29]. αKlotho’s ability to block this pathway contributes to its anti-tumor properties [30]. Transforming growth factor beta (TGFβ) is also able to modulate cell differentiation and mobility, namely inducing epithelial-to-mesenchymal transition [31], and αKlotho can block those effects [32]. The signaling pathways modulated by γKlotho are less studied, but Hori et al. (2016) implicated this protein in epithelial-to-mesenchymal transition in bladder cancer.
Accumulating data suggests that the tissue expression of Klotho proteins and, especially, the detection and quantitation of their soluble forms in body fluids like blood serum may be useful for establishing the diagnosis and prognosis of some types of cancer [6,33,34]. The present review aims to analyze scientific data regarding the role of Klotho proteins in cancer and to retrieve information regarding their potential use as diagnostic and prognostic biomarkers.

2. Materials and Methods

The review was performed on three standard databases on biomedicine: PubMed, Scielo, and ScienceDirect, accessed in April 2023, including scientific papers published between 2012 and December 2022. The keywords “cancer AND Klotho” were applied. The following inclusion criteria were established concerning the type of study (case series and case–control studies in humans; experimental in vitro and in vivo studies) and outcomes (effects of Klotho gene products in cancer). Exclusion criteria were lack of clear definition of cancer type or controls, lack of Klotho gene product quantification, case reports, review articles, commentaries, hypothesis and meta-analyses, and languages other than English. The abstracts and, when necessary, the materials and methods were analyzed to apply inclusion and exclusion criteria (Figure 2).

3. Results

Most publications were excluded due to duplication between databases or by applying exclusion criteria. Many articles have dealt with other pathologies where Klotho proteins are thought to play significant roles, most prominently in renal diseases. Overall, after applying inclusion and exclusion criteria, 26 articles were selected for further analysis (Table 1). Most studies used in vitro and/or clinical observational approaches, with only 7 articles using in vivo studies with animal models. Clinical observational studies often described the expression of Klotho genes at the RNA and/or protein levels and provided correlations between these markers’ expression levels and relevant clinical parameters. Caseloads varied between 36 and 313 patients. Remarkably, none of the clinical studies adopted an interventional approach, and most consisted of retrospective cohort studies, while one article included a case–control study. In vitro studies provided insights into the regulation of Klotho protein’s expression and its effects on cancer cells. Among the 26 selected articles, 21 dealt with αKlotho, 5 with βKlotho and only 3 with γKlotho, with one article studying α and βKlotho and another studying all the three proteins.

3.1. αKlotho

The main findings of the 21 articles addressing αKlotho in cancer are summarized in Table 2. Four studies were focused on colorectal cancer [46,48,50,53], another three on lung cancer [35,42,45], two on hepatocellular carcinoma [30,36], two on ovarian cancer [38,43], two on renal cell carcinoma [28,37], and two on gastric cancer [49,52]. Prostate cancer [27], acute myeloid leukemia [39], thyroid cancer [40], esophageal cancer [41], breast cancer [21], and pancreatic cancer [51] were each studied by a single article.

3.1.1. Clinicopathological Characteristics

αKlotho was generally found to act as a tumor suppressor, and its downregulation was consistently associated with aggressive tumor phenotypes and worse prognosis. In prostate cancer, αKlotho protein expression was detected in 50% of primary and 90% of metastatic samples [27]. In lung cancer, αKlotho was detected in most samples, but its expression pattern seems to be subtype-specific and requires further studies [40,45]. In hepatocellular carcinoma, αKlotho tissue expression is downregulated in tumor versus adjacent tissues and inversely correlates with tumor size, TNM stage, and nuclear grade [36]. Similar findings were obtained when studying renal cell carcinoma [30,39]. In breast [21], esophageal [41] and ovarian [38,43] cancer, αKlotho expression is downregulated compared with normal tissues.

3.1.2. Diagnosis

Soluble αKlotho can be quantified in blood serum using ELISA, and αKlotho levels were also suggested to have diagnostic value for esophageal cancer [41].

3.1.3. Survival and Treatment Response

The quantitation of αKlotho expression levels on tumor tissues using immunohistochemistry (IHC) was of prognostic significance in colorectal, esophageal, hepatocellular, lung, and ovarian cancer [36,41,43,45,46]. KLA promoter methylation and mRNA expression levels by quantitative real-time PCR (qRT-PCR) were also reported to have prognostic value in hepatocellular carcinoma and pancreatic cancer [36,51]. Reduced αKlotho serum levels were associated with reduced cancer-specific survival and progression-free survival among renal cell carcinoma patients [37]. Interestingly, reduced αKlotho levels were also suggested to promote cytarabine resistance in acute myeloid leukemia cells [39].

3.2. βKlotho

The 5 articles focused on βKlotho are addressed in Table 3, which summarizes their main findings. Two articles dealt with hepatocellular carcinoma [26,54], while prostate cancer [27], breast cancer [21], and pancreatic adenocarcinoma [44] were studied in one article each.

3.2.1. Clinicopathological Characteristics

In hepatocellular carcinoma, βKlotho was proposed to mediate tumor aggressiveness via FGFR signaling [26,54]. Conversely, in breast and pancreatic cancers, βKlotho was proposed to act as a tumor suppressor [21,44]. In prostate cancer, βKlotho protein expression was detected in a majority of primary and metastatic lesions [27].

3.2.2. Survival and Treatment Response

Interestingly, one study on hepatocellular carcinoma [26] showed that a >2-fold increase in KLB gene expression correlates with the development of multiple versus single lesions. A pre-clinical study [55] suggested that βKlotho mediates FGF9 pro-survival functions in hepatocellular carcinoma via FGFR3 and FGFR4 and may be useful in selecting patients who could benefit from anti-FGFR therapies. A similar scenario was suggested by a single study focused on prostate cancer [27].

3.3. γKlotho

γKlotho was studied in three articles, summarized in Table 4. Breast [21], prostate [47], and bladder cancers [22] were studied in one article each. All three articles found that higher γKlotho expression is associated with cancer aggressiveness and poor prognosis, suggesting that γKlotho levels assessed at the mRNA or the protein level may be useful to predict patient survival and response to therapy.

3.3.1. Clinicopathological Characteristics

Triple-negative breast cancer is an aggressive breast cancer subtype that poses a significant therapeutic challenge [56]. LCTL gene expression was found to be upregulated in triple-negative breast cancer samples, and expression levels correlated with increased cell proliferation, histological grade, and TNM stage [21]. Bladder cancer includes muscle-invasive and non-muscle-invasive forms [57] with distinct biological behavior. Higher γKlotho protein expression was observed in muscle-invasive versus non-muscle-invasive lesions [22].

3.3.2. Survival and Treatment Response

In triple-negative breast cancer, LCTL gene expression levels correlated with reduced progression-free survival [21]. Castration-resistant prostate cancer is another challenging malignancy with heterogeneous morphological and molecular phenotypes [55,58]. High γKlotho expression levels, as demonstrated by IHC, were shown to correlate with reduced overall survival and poor response to docetaxel in patients and in a mouse xenograft model [40]. In non-muscle-invasive bladder cancer, γKlotho protein levels were shown to correlate with reduced progression-free survival [22].

4. Discussion

The three Klotho proteins have complex roles in different types of cancer. The role of γKlotho is less well defined than that of its related Klotho proteins, partially because of its unusual molecular structure and because it was discovered more recently. The present review organized data from scientific articles published between 2012 and 2022 regarding the roles of Klotho proteins in cancer and their potential use as diagnostic and prognostic tools.
The role of all three proteins was studied in prostate cancer. This is a highly prevalent disease in middle-aged to older men that usually develops as an androgen-dependent adenocarcinoma but may progress to an androgen-independent castration-resistant phenotype and small-cell neoplasia, often displaying neuroendocrine markers, which are associated with poor patient prognosis [55]. αKlotho and βKlotho expression was detected in prostate cancer cell lines representing prostate adenocarcinoma and small-cell carcinoma, as well as in tumor tissues from primary tumors and metastasis, where they seem to mediate FGFR signaling [27]. It was further suggested that IHC tests for detecting αKlotho and βKlotho in tumor tissue may be of use to predict response to anti-FGFR therapies [27]. γKlotho expression in castration-resistant prostate cancer was associated with reduced survival and resistance to docetaxel [47], which is used as chemotherapy for such advanced cases [59]. Taken together, these results suggest that the immuno-expression patterns of Klotho proteins on prostate cancer tissues may be a valuable tool for tailoring treatment regimens for specific patients.
Lung cancer is also a common and aggressive malignancy, which includes multiple subtypes with distinct biological behavior [60]. Loss of αKlotho expression was consistently associated with increased tumor aggressiveness in three studies using in vitro and in vivo models [35] and clinical observational studies of neuroendocrine tumors [45], early centrally located cancers, and squamous cell carcinomas [42]. The observation that αKlotho may predict survival in patients with large cell neuroendocrine lung cancer is of particular interest, as it suggests that this marker has prognostic value in this specific lung cancer subtype [45]. Additionally, limited in vivo and in vitro data suggest that αKlotho downregulation may predict resistance to cisplatin-based chemotherapy [35], but additional studies are required to confirm this hypothesis.
Hepatocellular carcinoma is the most common type of liver cancer [61]. Although αKlotho was reported to act as a tumor suppressor [30,36], βKlotho showed oncogenic activity via enhanced FGFR signaling [26,54]. Importantly, αKlotho gene promoter methylation and protein expression may be of use as prognostic markers to estimate patient survival [36], while βKlotho may be a useful marker to predict response to anti-FGFR therapies [26].
In renal cell carcinoma, αKlotho downregulation was also reported to act as a tumor suppressor, and its loss was associated with tumor aggressiveness [28,42]. Of particular interest is the use of ELISA tests to detect soluble αKlotho in blood serum samples, as reduced levels of this protein were significantly associated with patients with the clear cell subtype of RCC [37]. These findings suggest that such tests may be used in liquid biopsies to help establish the prognosis of specific RCC patient subgroups.
Ovarian cancer is a frequent malignancy in women [62], and αKlotho was reported to act as a tumor suppressor in this type of cancer using experimental and clinical approaches [38,44]. Importantly, one study suggested that reduced αKlotho immuno-expression in cancer tissues may be useful as a prognostic marker to predict poor patient survival [44]. The same study reported that αKlotho was associated with higher interleukin-6 (IL-6) circulating levels. IL-6 is a pro-inflammatory cytokine that mediates some paraneoplastic syndromes like cancer cachexia [63], so it is interesting to speculate that αKlotho expression levels may also be used to predict the development of such syndromes.
In acute myeloid leukemia, loss of αKlotho was reported to be associated with cytarabine resistance in vitro, suggesting its possible use as a tool to design tailored therapies for leukemia patients [39]. Additional studies are needed to test this hypothesis, as cytarabine remains an important drug for treating this type of leukemia [64].
Breast cancer is highly prevalent in women and is often life-threatening [56]. In one study, αKlotho and βKlotho were downregulated in tumor tissue versus adjacent tissue, suggesting they act as tumor suppressors [21]. Conversely, higher γKlotho (LCTL) gene expression levels using qRT-PCR were found in cancer versus adjacent tissue, specifically in the aggressive triple-negative cancer subtype [21,65], suggesting it is associated with tumor aggressiveness. Interestingly, it was suggested that qRT-PCR for LCTL may be useful as a prognostic marker to estimate patient survival in patients with triple-negative breast cancer [21].
In papillary thyroid cancer, a single study [40] reported that αKlotho was able to reduce cell proliferation and induce apoptosis in vitro. The potential use of this protein for diagnostic and prognostic purposes in thyroid cancer remains to be determined.
In esophageal cancer, an interesting study [41] reported that the levels of soluble αKlotho in blood serum as detected by ELISA were higher in patients versus healthy controls. A cut-off value was estimated that allowed researchers to distinguish between patients and controls with approximately 81% sensitivity and specificity. Interestingly, in tissue samples, αKlotho was expressed at higher levels in adjacent versus tumor samples, and αKlotho downregulation correlated with increased tumor aggressiveness and reduced patient survival. These data highlight the potential of αKlotho as a marker in liquid biopsies for the diagnosis of esophageal cancer, while tissue levels may have prognostic significance.
Colorectal cancer is highly prevalent in multiple world regions, and large bowel carcinogenesis is associated with chronic inflammation [66]. In this type of cancer, 4 studies consistently reported that αKlotho acts as a tumor suppressor [46,48,50,53]. In vitro tests revealed new regulatory pathways that control αKlotho expression via FL-1 [48] and support the pro-apoptotic role of αKlotho via TRAIL [50]. Interestingly, one study described how αKlotho downregulation promotes a senescence-associated secretory phenotype in mesenchymal cells that may contribute to tumorigenesis via the nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) signaling pathway [46]. This is a pivotal mediator of inflammation and tissue repair, but also of carcinogenesis in specific settings. Chronic inflammation is a key player in colon cancer, and the secretion of NFκB-controlled C-C motif chemokine ligand 2 (CCL2) by senescent stromal cells was proposed to promote carcinogenesis of the colon. αKlotho abrogated CCL2 signaling and was associated with improved patient survival, suggesting it may be of use as a prognostic marker.
Two in vitro studies addressed the role of αKlotho in gastric cancer, further associating αKlotho downregulation with aggressive cancer phenotypes [49,52]. SOX17 and an epigenetic pathway involving circular RNA ITCH and miR-199-5p were shown to regulate αKlotho expression in gastric cancer cells. Although these findings support the role of αKlotho as a tumor suppressor, further developments are needed to explore its potential role as a diagnostic or prognostic marker in gastric cancer.
A single study addressed the role of αKlotho in pancreatic adenocarcinoma and concluded that KLA gene expression levels and promoter methylation may have prognostic value, as increased KLA promoter methylation and decreased mRNA expression levels were associated with lower patient survival [51]. This was further supported by tests in three complementary mouse models, where αKlotho decreased cancer growth and improved survival. Another study using expression data from the GEO database also suggested that KLB upregulation is associated with improved survival in pancreatic cancer patients [46]. Taken together, these data provide evidence to support the further development of Klotho as a prognostic marker in pancreatic adenocarcinoma.
Urothelial carcinoma of the urinary bladder is a common malignancy that includes highly aggressive forms that invade the bladder’s muscular layer and non-muscle-invasive forms associated with local recurrence [67]. One study reported that γKlotho expression was observed in both muscle-invasive and non-muscle-invasive bladder cancer using IHC and that expression levels were associated with poor overall survival among patients with non-muscle-invasive cancer [22].

5. Conclusions

Overall, the datasets published between 2012 and 2022 provide evidence supporting the development of Klotho genes and their mRNA and protein products as potential prognostic markers in multiple types of cancer, especially in the prediction of patient survival. Although αKlotho was consistently associated with improved patient prognosis, γKlotho was associated with increased cancer aggressiveness, and βKlotho showed mixed results. It is critical to accurately identify specific tumor subtypes where Klotho is of interest (muscle-invasive versus non-muscle-invasive urothelial carcinoma) to take the most advantage of its potential. The use of Klotho levels as diagnostic markers was less frequently observed in the literature, although one study provided detailed data regarding soluble αKlotho levels in blood serum and the diagnosis of esophageal cancer. However, most studies still did not present such detailed results, and the clinical use of Klotho will require additional development.

Author Contributions

Conceptualization, R.M.G.d.C. and H.O.B.; methodology, M.S.d.A. and A.M.M.L.; validation, F.F.L. and J.I.S.G., investigation, J.M., M.M.A.L.S. and J.I.S.G.; resources, A.I.F.-R., P.A.O. and R.M.G.d.C.; data curation, M.S.d.A., M.M.A.L.S. and F.F.L.; writing—original draft preparation, R.M.G.d.C. and A.I.F.-R.; writing—review and editing, H.O.B., M.S.d.A., F.F.L., M.M.A.L.S. and P.A.O.; supervision, F.F.L. and R.M.G.d.C.; funding acquisition, M.S.d.A., P.A.O. and H.O.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by CAPES (grant numbers 001, 0810/2020/88881.510244/2020-01), FAPEMA (IECT-FAPEMA-05796/18, FAPEMA IECT 30/2018—IECT Saúde, PPSUS-02160/20, BPD-01343/23), CI-IPOP (PI86-CI-IPOP-66-2017), FCT (UID/AGR/04033/2020, UIDB/CVT/00772/2020, LA/P/0045/2020, UIDB/00511/2020, UIDP/00511/2020, NORTE-01-0145-FEDER-000054).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data produced in this study are available in this article.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study, in the collection, analyses, or interpretation of data, in the writing of the manuscript, or in the decision to publish the results.

References

  1. 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]
  2. Matsumuraab, Y.; Aizawaab, H.; Shiraki-Iida, T.; Nagaibd, R.; Kuro-O, M.; Nabeshima, Y.-I. Identification of the HumanKlothoGene and Its Two Transcripts Encoding Membrane and SecretedKlothoProtein. Biochem. Biophys. Res. Commun. 1998, 242, 626–630. [Google Scholar] [CrossRef] [PubMed]
  3. Ito, S.; Kinoshita, S.; Shiraishi, N.; Nakagawa, S.; Sekine, S.; Fujimori, T.; Nabeshima, Y.-I. Molecular cloning and expression analyses of mouse betaβklotho, which encodes a novel Klotho family protein. Mech. Dev. 2000, 98, 115–119. [Google Scholar] [CrossRef] [PubMed]
  4. Chen, C.-D.; Podvin, S.; Gillespie, E.; Leeman, S.E.; Abraham, C.R. Insulin stimulates the cleavage and release of the extracellular domain of Klotho by ADAM10 and ADAM17. Proc. Natl. Acad. Sci. USA 2007, 104, 19796–19801. [Google Scholar] [CrossRef]
  5. Kuro-O, M. The Klotho proteins in health and disease. Nat. Rev. Nephrol. 2019, 15, 27–44. [Google Scholar] [CrossRef]
  6. Bednarska, S.; Fryczak, J.; Siejka, A. Serum β-Klotho concentrations are increased in women with polycystic ovary syndrome. Cytokine 2020, 134, 155188. [Google Scholar] [CrossRef]
  7. Ito, S.; Fujimori, T.; Hayashizaki, Y.; Nabeshima, Y.-I. Identification of a novel mouse membrane-bound family 1 glycosidase-like protein, which carries an atypical active site structure. Biochim. Biophys. Acta (BBA)-Gene Struct. Expr. 2002, 1576, 341–345. [Google Scholar] [CrossRef]
  8. Kurosu, H.; Ogawa, Y.; Miyoshi, M.; Yamamoto, M.; Nandi, A.; Rosenblatt, K.P.; Baum, M.G.; Schiavi, S.; Hu, M.-C.; Moe, O.W.; et al. Regulation of Fibroblast Growth Factor-23 Signaling by Klotho. J. Biol. Chem. 2006, 281, 6120–6123. [Google Scholar] [CrossRef]
  9. Urakawa, I.; Yamazaki, Y.; Shimada, T.; Iijima, K.; Hasegawa, H.; Okawa, K.; Fujita, T.; Fukumoto, S.; Yamashita, T. Klotho converts canonical FGF receptor into a specific receptor for FGF23. Nature 2006, 444, 770–774. [Google Scholar] [CrossRef]
  10. Lee, S.; Choi, J.; Mohanty, J.; Sousa, L.P.; Tome, F.; Pardon, E.; Steyaert, J.; Lemmon, M.A.; Lax, I.; Schlessinger, J. Structures of β-klotho reveal a ‘zip code’-like mechanism for endocrine FGF signalling. Nature 2018, 553, 501–505. [Google Scholar] [CrossRef]
  11. Lin, B.C.; Wang, M.; Blackmore, C.; Desnoyers, L.R. Liver-specific Activities of FGF19 Require Klotho beta. J. Biol. Chem. 2007, 282, 27277–27284. [Google Scholar] [CrossRef] [PubMed]
  12. Ogawa, Y.; Kurosu, H.; Yamamoto, M.; Nandi, A.; Rosenblatt, K.P.; Goetz, R.; Eliseenkova, A.V.; Mohammadi, M.; Kuro-O, M. βKlotho is required for metabolic activity of fibroblast growth factor 21. Proc. Natl. Acad. Sci. USA 2007, 104, 7432–7437. [Google Scholar] [CrossRef] [PubMed]
  13. Kurosu, H.; Choi, M.; Ogawa, Y.; Dickson, A.S.; Goetz, R.; Eliseenkova, A.V.; Mohammadi, M.; Rosenblatt, K.P.; Kliewer, S.A.; Kuro-O, M. Tissue-specific Expression of βKlotho and Fibroblast Growth Factor (FGF) Receptor Isoforms Determines Metabolic Activity of FGF19 and FGF21. J. Biol. Chem. 2007, 282, 26687–26695. [Google Scholar] [CrossRef] [PubMed]
  14. Kharitonenkov, A.; Dunbar, J.D.; Bina, H.A.; Bright, S.; Moyers, J.S.; Zhang, C.; Ding, L.; Micanovic, R.; Mehrbod, S.F.; Knierman, M.D.; et al. FGF-21/FGF-21 receptor interaction and activation is determined by βKlotho. J. Cell. Physiol. 2007, 215, 1–7. [Google Scholar] [CrossRef]
  15. Brownstein, C.A.; Adler, F.; Nelson-Williams, C.; Iijima, J.; Li, P.; Imura, A.; Nabeshima, Y.-I.; Reyes-Mugica, M.; Carpenter, T.O.; Lifton, R.P. A translocation causing increased α-Klotho level results in hypophosphatemic rickets and hyperparathyroidism. Proc. Natl. Acad. Sci. USA 2008, 105, 3455–3460. [Google Scholar] [CrossRef]
  16. Stenvinkel, P.; Painer, J.; Kuro-O, M.; Lanaspa, M.; Arnold, W.; Ruf, T.; Shiels, P.G.; Johnson, R.J. Novel treatment strategies for chronic kidney disease: Insights from the animal kingdom. Nat. Rev. Nephrol. 2018, 14, 265–284. [Google Scholar] [CrossRef]
  17. Ligumsky, H.; Merenbakh-Lamin, K.; Keren-Khadmy, N.; Wolf, I.; Rubinek, T. The role of α-klotho in human cancer: Molecular and clinical aspects. Oncogene 2022, 41, 4487–4497. [Google Scholar] [CrossRef]
  18. Ye, X.; Guo, Y.; Zhang, Q.; Chen, W.; Hua, X.; Liu, W.; Yang, Y.; Chen, G. βKlotho Suppresses Tumor Growth in Hepatocellular Carcinoma by Regulating Akt/GSK-3β/Cyclin D1 Signaling Pathway. PLoS ONE 2013, 8, e55615. [Google Scholar] [CrossRef]
  19. Liu, Z.; Qi, S.; Zhao, X.; Li, M.; Ding, S.; Lu, J.; Zhang, H. Metformin inhibits 17β-estradiol-induced epithelial-to-mesenchymal transition via βKlotho-related ERK1/2 signaling and AMPKα signaling in endometrial adenocarcinoma cells. Oncotarget 2016, 7, 21315–21331. [Google Scholar] [CrossRef]
  20. Cui, G.; Martin, R.C.; Jin, H.; Liu, X.; Pandit, H.; Zhao, H.; Cai, L.; Zhang, P.; Li, W.; Li, Y. Up-regulation of FGF15/19 signaling promotes hepatocellular carcinoma in the background of fatty liver. J. Exp. Clin. Cancer Res. 2018, 37, 136. [Google Scholar] [CrossRef]
  21. Trošt, N.; Peña-Llopis, S.; Koirala, S.; Stojan, J.; Potts, P.R.; Tacer, K.F.; Martinez, E.D. γKlotho is a novel marker and cell survival factor in a subset of triple negative breast cancers. Oncotarget 2016, 7, 2611–2628. [Google Scholar] [CrossRef] [PubMed]
  22. Hori, S.; Miyake, M.; Tatsumi, Y.; Morizawa, Y.; Nakai, Y.; Onishi, S.; Onishi, K.; Iida, K.; Gotoh, D.; Tanaka, N.; et al. Gamma-Klotho exhibits multiple roles in tumor growth of human bladder cancer. Oncotarget 2018, 9, 19508–19524. [Google Scholar] [CrossRef] [PubMed]
  23. Rubinek, T.; Wolf, I. The Role of Alpha-Klotho as a Universal Tumor Suppressor. Vitam. Horm. 2016, 101, 197–214. [Google Scholar] [CrossRef]
  24. 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] [PubMed]
  25. Sachdeva, A.; Gouge, J.; Kontovounisios, C.; Nikolaou, S.; Ashworth, A.; Lim, K.; Chong, I. Klotho and the Treatment of Human Malignancies. Cancers 2020, 12, 1665. [Google Scholar] [CrossRef]
  26. Poh, W.; Wong, W.; Ong, H.; Aung, M.O.; Lim, S.G.; Chua, B.T.; Ho, H.K. Klotho-beta overexpression as a novel target for suppressing proliferation and fibroblast growth factor receptor-4 signaling in hepatocellular carcinoma. Mol. Cancer 2012, 11, 14. [Google Scholar] [CrossRef]
  27. Feng, S.; Dakhova, O.; Creighton, C.J.; Ittmann, M. Endocrine Fibroblast Growth Factor FGF19 Promotes Prostate Cancer Progression. Cancer Res. 2013, 73, 2551–2562. [Google Scholar] [CrossRef] [PubMed]
  28. Zhu, Y.; Xu, L.; Zhang, J.; Xu, W.; Liu, Y.; Yin, H.; Lv, T.; An, H.; Liu, L.; He, H.; et al. Klotho suppresses tumor progression via inhibiting PI3K/Akt/GSK3β/Snail signaling in renal cell carcinoma. Cancer Sci. 2013, 104, 663–671. [Google Scholar] [CrossRef]
  29. Suzuki, H.; Watkins, D.N.; Jair, K.-W.; Schuebel, K.E.; Markowitz, S.D.; Chen, W.D.; Pretlow, T.P.; Yang, B.; Akiyama, Y.; van Engeland, M.; et al. Epigenetic inactivation of SFRP genes allows constitutive WNT signaling in colorectal cancer. Nat. Genet. 2004, 36, 417–422. [Google Scholar] [CrossRef] [PubMed]
  30. Sun, H.; Gao, Y.; Lu, K.; Zhao, G.; Li, X.; Li, Z.; Chang, H. Overexpression of Klotho suppresses liver cancer progression and induces cell apoptosis by negatively regulating wnt/β-catenin signaling pathway. World J. Surg. Oncol. 2015, 13, 307. [Google Scholar] [CrossRef] [PubMed]
  31. Pickup, M.; Novitskiy, S.; Moses, H.L. The roles of TGFβ in the tumour microenvironment. Nat. Rev. Cancer 2013, 13, 788–799. [Google Scholar] [CrossRef]
  32. Doi, S.; Zou, Y.; Togao, O.; Pastor, J.V.; John, G.B.; Wang, L.; Shiizaki, K.; Gotschall, R.; Schiavi, S.; Yorioka, N.; et al. Klotho Inhibits Transforming Growth Factor-β1 (TGF-β1) Signaling and Suppresses Renal Fibrosis and Cancer Metastasis in Mice. J. Biol. Chem. 2011, 286, 8655–8665. [Google Scholar] [CrossRef] [PubMed]
  33. Hori, S.; Miyake, M.; Onishi, S.; Tatsumi, Y.; Morizawa, Y.; Nakai, Y.; Anai, S.; Tanaka, N.; Fujimoto, K. Clinical significance of α- and β-Klotho in urothelial carcinoma of the bladder. Oncol. Rep. 2016, 36, 2117–2125. [Google Scholar] [CrossRef] [PubMed]
  34. Zhou, J.; Ben, S.; Xu, T.; Xu, L.; Yao, X. Serum β-klotho is a potential biomarker in the prediction of clinical outcomes among patients with NSCLC. J. Thorac. Dis. 2021, 13, 3137–3150. [Google Scholar] [CrossRef] [PubMed]
  35. Wang, Y.; Chen, L.; Huang, G.; He, D.; He, J.; Xu, W.; Zou, C.; Zong, F.; Li, Y.; Chen, B.; et al. Klotho Sensitizes Human Lung Cancer Cell Line to Cisplatin via PI3k/Akt Pathway. PLoS ONE 2013, 8, e57391. [Google Scholar] [CrossRef] [PubMed]
  36. Xie, B.; Zhou, J.; Yuan, L.; Ren, F.; Liu, D.-C.; Li, Q.; Shu, G. Epigenetic silencing of Klotho expression correlates with poor prognosis of human hepatocellular carcinoma. Hum. Pathol. 2013, 44, 795–801. [Google Scholar] [CrossRef]
  37. Gigante, M.; Lucarelli, G.; Divella, C.; Netti, G.S.; Pontrelli, P.; Cafiero, C.; Grandaliano, G.; Castellano, G.; Rutigliano, M.; Stallone, G.; et al. Soluble Serum αKlotho Is a Potential Predictive Marker of Disease Progression in Clear Cell Renal Cell Carcinoma. Medicine 2015, 94, e1917. [Google Scholar] [CrossRef]
  38. Lojkin, I.; Rubinek, T.; Orsulic, S.; Schwarzmann, O.; Karlan, B.Y.; Bose, S.; Wolf, I. Reduced expression and growth inhibitory activity of the aging suppressor klotho in epithelial ovarian cancer. Cancer Lett. 2015, 362, 149–157. [Google Scholar] [CrossRef]
  39. Shibayama, Y.; Kondo, T.; Ohya, H.; Fujisawa, S.-I.; Teshima, T.; Iseki, K. Upregulation of microRNA-126-5p is associated with drug resistance to cytarabine and poor prognosis in AML patients. Oncol. Rep. 2015, 33, 2176–2182. [Google Scholar] [CrossRef]
  40. Dai, D.; Wang, Q.; Li, X.; Liu, J.; Ma, X.; Xu, W. Klotho inhibits human follicular thyroid cancer cell growth and promotes apoptosis through regulation of the expression of stanniocalcin-1. Oncol. Rep. 2015, 35, 552–558. [Google Scholar] [CrossRef]
  41. Tang, X.; Fan, Z.; Wang, Y.; Ji, G.; Wang, M.; Lin, J.; Huang, S. Expression of klotho and β-catenin in esophageal squamous cell carcinoma, and their clinicopathological and prognostic significance. Dis. Esophagus 2016, 29, 207–214. [Google Scholar] [CrossRef] [PubMed]
  42. Ibi, T.; Usuda, J.; Inoue, T.; Sato, A.; Takegahara, K. Klotho expression is correlated to molecules associated with epithelial-mesenchymal transition in lung squamous cell carcinoma. Oncol. Lett. 2017, 14, 5526–5532. [Google Scholar] [CrossRef] [PubMed]
  43. Yan, Y.; Wang, Y.; Xiong, Y.; Lin, X.; Zhou, P.; Chen, Z. Reduced Klotho expression contributes to poor survival rates in human patients with ovarian cancer, and overexpression of Klotho inhibits the progression of ovarian cancer partly via the inhibition of systemic inflammation in nude mice. Mol. Med. Rep. 2017, 15, 1777–1785. [Google Scholar] [CrossRef] [PubMed]
  44. Haq, F.; Sung, Y.-N.; Park, I.; Kayani, M.A.; Yousuf, F.; Hong, S.-M.; Ahn, S.-M. FGFR1 expression defines clinically distinct subtypes in pancreatic cancer. J. Transl. Med. 2018, 16, 374. [Google Scholar] [CrossRef] [PubMed]
  45. Brominska, B.; Gabryel, P.; Jarmołowska-Jurczyszyn, D.; Janicka-Jedyńska, M.; Kluk, A.; Trojanowski, M.; Brajer-Luftmann, B.; Woliński, K.; Czepczyński, R.; Gut, P.; et al. Klotho expression and nodal involvement as predictive factors for large cell lung carcinoma. Arch. Med. Sci. 2019, 15, 1010–1016. [Google Scholar] [CrossRef] [PubMed]
  46. Liu, Y.; Pan, J.; Pan, X.; Wu, L.; Bian, J.; Lin, Z.; Xue, M.; Su, T.; Lai, S.; Chen, F.; et al. Klotho-mediated targeting of CCL 2 suppresses the induction of colorectal cancer progression by stromal cell senescent microenvironments. Mol. Oncol. 2019, 13, 2460–2475. [Google Scholar] [CrossRef]
  47. Onishi, K.; Miyake, M.; Hori, S.; Onishi, S.; Iida, K.; Morizawa, Y.; Tatsumi, Y.; Nakai, Y.; Tanaka, N.; Fujimoto, K. γ-Klotho is correlated with resistance to docetaxel in castration-resistant prostate cancer. Oncol. Lett. 2020, 19, 2306–2316. [Google Scholar] [CrossRef]
  48. Xie, B.; Hu, F.; Li, M.; Mo, L.; Xu, C.; Xiao, Y.; Wang, X.; Nie, J.; Yang, L.; He, Y. FLI-1 mediates tumor suppressor function via Klotho signaling in regulating CRC. Cell Biol. Int. 2020, 44, 1514–1522. [Google Scholar] [CrossRef]
  49. Yang, L.; Wu, Y.; He, H.; Hu, F.; Li, M.; Mo, L.; Xiao, Y.; Wang, X.; Xie, B. Delivery of BR2-SOX17 fusion protein can inhibit cell survival, proliferation, and invasion in gastric cancer cells through regulating Klotho gene expression. Cell Biol. Int. 2020, 44, 2011–2020. [Google Scholar] [CrossRef]
  50. GGunes, S.; Soykan, M.N.; Sariboyaci, A.E.; Uysal, O.; Sevimli, T.S. Enhancement of Apo2L/TRAIL signaling pathway receptors by the activation of Klotho gene with CRISPR/Cas9 in Caco-2 colon cancer cells. Med. Oncol. 2021, 38, 146. [Google Scholar] [CrossRef]
  51. Rubinstein, T.A.; Reuveni, I.; Hesin, A.; Klein-Goldberg, A.; Olauson, H.; Larsson, T.E.; Abraham, C.R.; Zeldich, E.; Bosch, A.; Chillón, M.; et al. A Transgenic Model Reveals the Role of Klotho in Pancreatic Cancer Development and Paves the Way for New Klotho-Based Therapy. Cancers 2021, 13, 6297. [Google Scholar] [CrossRef] [PubMed]
  52. Wang, Y.; Wang, H.; Zheng, R.; Wu, P.; Sun, Z.; Chen, J.; Zhang, L.; Zhang, C.; Qian, H.; Jiang, J.; et al. Circular RNA ITCH suppresses metastasis of gastric cancer via regulating miR-199a-5p/Klotho axis. Cell Cycle 2021, 20, 522–536. [Google Scholar] [CrossRef] [PubMed]
  53. Sariboyaci, A.E.; Uysal, O.; Soykan, M.N.; Gunes, S. The potential therapeutic effect of klotho on cell viability in human colorectal adenocarcinoma HT-29 cells. Med. Oncol. 2022, 39, 191. [Google Scholar] [CrossRef] [PubMed]
  54. Tao, Z.; Cui, Y.; Xu, X.; Han, T. FGFR redundancy limits the efficacy of FGFR4-selective inhibitors in hepatocellular carcinoma. Proc. Natl. Acad. Sci. USA 2022, 119, e2208844119. [Google Scholar] [CrossRef]
  55. Labrecque, M.P.; Coleman, I.M.; Brown, L.G.; True, L.D.; Kollath, L.; Lakely, B.; Nguyen, H.M.; Yang, Y.C.; Gil da Costa, R.M.; Kaipainen, A.; et al. Molecular profiling stratifies diverse phenotypes of treatment-refractory metastatic castration-resistant prostate cancer. J. Clin. Investig. 2019, 129, 4492–4505. [Google Scholar] [CrossRef]
  56. Harbeck, N.; Gnant, M. Breast cancer. Lancet 2017, 389, 1134–1150. [Google Scholar] [CrossRef]
  57. Gil da Costa, R.M.; Levesque, C.; Bianchi-Frias, D.; Chatterjee, P.; Lam, H.; Santos, C.; Coleman, I.M.; Ferreirinha, P.; Vilanova, M.; da Cunha, N.P.; et al. Pharmacological NF-κB inhibition decreases cisplatin chemoresistance in muscle-invasive bladder cancer and reduces cisplatin-induced toxicities. Mol. Oncol. 2023. [Google Scholar] [CrossRef]
  58. DeLucia, D.C.; Cardillo, T.M.; Ang, L.S.; Labrecque, M.P.; Zhang, A.; Hopkins, J.E.; De Sarkar, N.; Coleman, I.; Gil da Costa, R.M.; Corey, E.; et al. Regulation of CEACAM5 and Therapeutic Efficacy of an Anti-CEACAM5–SN38 Antibody–drug Conjugate in Neuroendocrine Prostate Cancer. Clin. Cancer Res. 2021, 27, 759–774. [Google Scholar] [CrossRef]
  59. Tannock, I.F.; De Wit, R.; Berry, W.R.; Horti, J.; Pluzanska, A.; Chi, K.N.; Oudard, S.; Théodore, C.; James, N.D.; Turesson, I.; et al. Docetaxel plus Prednisone or Mitoxantrone plus Prednisone for Advanced Prostate Cancer. N. Engl. J. Med. 2004, 351, 1502–1512. [Google Scholar] [CrossRef]
  60. Rodriguez-Canales, J.; Parra-Cuentas, E.; Wistuba, I.I. Diagnosis and Molecular Classification of Lung Cancer. Cancer Treat. Res. 2016, 170, 25–46. [Google Scholar] [CrossRef]
  61. Forner, A.; Reig, M.; Bruix, J. Hepatocellular carcinoma. Lancet 2018, 391, 1301–1314. [Google Scholar] [CrossRef] [PubMed]
  62. Lheureux, S.; Braunstein, M.; Oza, A.M. Epithelial ovarian cancer: Evolution of management in the era of precision medicine. CA A Cancer J. Clin. 2019, 69, 280–304. [Google Scholar] [CrossRef] [PubMed]
  63. Santos, J.M.O.; Costa, A.C.; Dias, T.R.; Satari, S.; e Silva, M.P.C.; Gil da Costa, R.M.; Medeiros, R. Towards Drug Repurposing in Cancer Cachexia: Potential Targets and Candidates. Pharmaceuticals 2021, 14, 1084. [Google Scholar] [CrossRef] [PubMed]
  64. Liu, H. Emerging agents and regimens for AML. J. Hematol. Oncol. 2021, 14, 49. [Google Scholar] [CrossRef]
  65. Bianchini, G.; De Angelis, C.; Licata, L.; Gianni, L. Treatment landscape of triple-negative breast cancer—Expanded options, evolving needs. Nat. Rev. Clin. Oncol. 2022, 19, 91–113. [Google Scholar] [CrossRef]
  66. Patel, S.G.; Karlitz, J.J.; Yen, T.; Lieu, C.H.; Boland, C.R. The rising tide of early-onset colorectal cancer: A comprehensive review of epidemiology, clinical features, biology, risk factors, prevention, and early detection. Lancet Gastroenterol. Hepatol. 2022, 7, 262–274. [Google Scholar] [CrossRef]
  67. Compérat, E.; Amin, M.B.; Cathomas, R.; Choudhury, A.; De Santis, M.; Kamat, A.; Stenzl, A.; Thoeny, H.C.; Witjes, J.A. Current best practice for bladder cancer: A narrative review of diagnostics and treatments. Lancet 2022, 400, 1712–1721. [Google Scholar] [CrossRef]
Figure 1. αKlotho downregulates signaling mediated by multiple cell membrane receptors, contributing to its anti-cancer effects.
Figure 1. αKlotho downregulates signaling mediated by multiple cell membrane receptors, contributing to its anti-cancer effects.
Diagnostics 13 03357 g001
Figure 2. Selection of articles from the PubMed, Scielo, and ScienceDirect databases and resulting publications for analysis.
Figure 2. Selection of articles from the PubMed, Scielo, and ScienceDirect databases and resulting publications for analysis.
Diagnostics 13 03357 g002
Table 1. Characteristics of the 26 articles included in the review.
Table 1. Characteristics of the 26 articles included in the review.
ReferenceYearType of CancerIn VitroIn VivoNumber of PatientsClinical
(Observational)
[26]2012Hepatocellular carcinomax 56Retrospective cohort
[27]2013Prostate cancerx 136Retrospective cohort
[35]2013Lung cancerxx -
[36]2013Hepatocellular carcinomax 64Retrospective cohort
[28]2013Renal cell carcinomax 125Retrospective cohort
[37]2015Renal cell carcinoma 160Retrospective cohort
[38]2015Ovarian cancerx 265Retrospective cohort
[39]2015Acute myeloid leukemiax 109Retrospective cohort
[30]2015Hepatocellular carcinomax -
[21]2015Breast cancer x 68Retrospective cohort
[40]2016Thyroid cancerx -
[41]2016Esophageal cancerx 160Retrospective case–control
[42]2017Pulmonary squamous cell carcinomax 40Retrospective cohort
[43]2017Ovarian cancerxx198Retrospective cohort
[44]2018Pancreatic
adenocarcinoma
x 313Retrospective cohort
[22]2018Bladder cancerxx205Retrospective cohort
[45]2019Large cell neuroendocrine lung cancer Retrospective cohort
[46]2019Colorectal cancerxx143Retrospective cohort
[47]2020Prostate cancer x36Retrospective cohort
[48]2020Colorectal cancerx -
[49]2020Gastric adenocarcinomax -
[50]2021Colorectal cancerx -
[51]2021Pancreatic cancer x178Retrospective cohort
[52]2021Gastric cancerx 94Retrospective cohort
[53]2022Colorectal cancer x -
[54]2022Hepatocellular carcinoma xx -
x denotes that in vivo and/or in vitro experiments were performed for each article.
Table 2. Studies dealing with αKlotho.
Table 2. Studies dealing with αKlotho.
Cancer TypeReferenceType of SampleMain FindingsPotential
Applications
Prostate cancer[27]Frozen and FFPE cancer tissues. PC3, DU145, VCaP, LNCaP cancer cell lines, PNT1a normal prostate cellsKLA gene expression detected in all cell lines by qRT-PCR and FGF19 stimulates PCa cells in vitro. αKlotho detected by IHC in 50% primary and 90% metastatic PCa samples Screening of patients who may benefit from anti-FGFR therapies and may be using IHC on tumor tissues
Lung cancer[35]A549 and H460 tumor cells and xenograftsαKlotho downregulation promotes cisplatin resistance in vitro and in vivo
[42]FFPE cancer tissues (centrally located early lung cancer and SCC), A549, and SQ5 tumor cell lines αKlotho expressed in 100% centrally located early lung cancer samples but only in 13% SCC using IHC. Inhibited N-cadherin expression in vitro
[45]FFPE cancer tissues (large cell neuroendocrine lung cancer)αKlotho expressed in ¾ patients and associated with survivalTissue expression may predict prognosis (survival)
Hepatocellular carcinoma[36]Frozen and FFPE tumor and adjacent tissues. HRPG2, BEL-7402, SMMC-7721, HL7702, HUH-7, MHCC-97-H cancer cell lines and L-02 hepatocytesαKlotho is downregulated at mRNA and protein levels in HCC versus adjacent tissue; promoter methylation and reduced protein expression correlate with reduced survivalαKlotho promoter methylation and protein expression may predict prognosis (survival)
[30]HepG2 and SMMC-7721 cancer cell lines, L-02 hepatocytesRecombinant αKlotho downregulates Wnt/β-catenin signaling, suppressing proliferation and inducing apoptosis
Renal cell
carcinoma
[28]786-O, OS-RC-2, ACHN, Caki-1 and Renca cancer cell lines. Tumor tissueαKlotho tissue expression (IHC) is inversely correlated with tumor size, TNM stage, and nuclear grade. In vitro blocked EMT via PI3K/Akt/GSK3 β/SnailPotential IHC marker of tumor aggressiveness
[37]Frozen tumor and adjacent tissue (clear cell RCC). Preoperative blood serumαKlotho is downregulated in tumor tissue at RNA (qRT-PCR) and protein (IHC) levels. Reduced serum levels (ELISA) associated with higher tumor volume, Fuhrman grade, clinical stage, reduced cancer-specific survival, and progression-free survival Serum αKlotho
levels using ELISA may predict prognosis, including survival.
Ovarian cancer[38]Tumor (high-grade papillary-serous adenocarcinoma) and adjacent ovarian tissues. 19 cancer cell lines αKlotho was reduced in tumor versus adjacent tissues (IHC) and in 16/19 cell lines (qRT-PCR)
[43]FFPE and frozen tumor and adjacent tissues. 7 cancer cell lines αKlotho was reduced in tumor versus adjacent tissues (IHC). Reduction correlates with low survival. Tumor xenografts expressing αKlotho had a smaller size. KLA−/− mice showed higher IL-6 levels in response to xenografts Tissue expression using IHC may predict survival
Acute myeloid leukemia[39]KG-1 cellsExposure to miR-126-5p decreased αKlotho levels and induced Akt phosphorylation and cytarabine resistance αKlotho may predict cytarabine resistance
Breast cancer[21]Frozen tumor and adjacent tissues. MDA-MB-231 and H357T cancer cell linesαKlotho was downregulated in cancer versus adjacent tissue. Undetectable in both cell lines
Follicular thyroid carcinoma[40]FTC133 and FTC238 cancer cell linesαKlotho reduced cell proliferation and induced apoptosis in vitro
Esophageal cancer[41]FFPE cancer and adjacent tissues. Blood serum from patients/controls αKlotho was downregulated in cancer versus adjacent tissue (IHC). Correlates with improved survival inversely correlated with staging, grade, lymph node metastasis, and β-catenin. Serum levels are higher in patients versus controls Tissue levels (IHC) may predict prognosis, including survival. Serum 327 pg/mL cut-off (ELISA) is diagnostic with a sensitivity of 81% and specificity of 81%
Colorectal cancer[46]FFPE tumor tissue. RKO and LoVo cancer cell lines, Wi-38, and HUVEC cells Lower αKlotho (IHC) is associated with lower patient survival. αKlotho prevents pro-tumorigenic effects of senescent cells in vitro and in vivo via NFκB/CCL2 blockadeTissue levels (IHC) may predict survival
[48]Six cancer cell lines and normal cells FL-1 regulates αKlotho expression in cancer cells
[50]CaCo-2 cellsαKlotho induces apoptosis via the TRAIL death receptor
[53]HT29 cancer cell line, CCD841 cellsαKlotho induces apoptosis specifically in cancer cells
Gastric cancer[49]6 cancer cell lines and normal cellsSOX17 regulates αKlotho expression in cancer cells in vitro
[52]HGC-27, AGS, MKN-45, MGC-803, HE-293-T cancer cell lines, GES-1 cellsCircular RNA ITCH upregulates αKlotho by sponging out miR-199-5p, inhibiting cell proliferation, migration, invasion, and EMT
Pancreatic cancer[51]TCGA pancreatic ductal adenocarcinoma datasets, 3 mouse modelsPromoter methylation and mRNA downregulation are associated with reduced survival. αKlotho knockdown synergized with Kras mutation to promote carcinogenesis. Soluble αKlotho inhibited xenograft growth and promoted the survival of KPC mice Methylation and expression levels may predict survival
Table 3. Studies dealing with βKlotho.
Table 3. Studies dealing with βKlotho.
Cancer TypeReferenceType of SampleMain FindingsPotential Applications
Hepatocellular carcinoma[26]Tumor and adjacent tissue in TrizolKLB gene expression is upregulated in cancer tissues. A >2-fold increase correlates with the development of multiple lesions.Screening of patients who could benefit from anti-FGFR therapies. Prediction of lesion multiplicity.
[54]Cell lines and xenograft mouse modelβKlotho mediates FGF9 pro-survival functions via FGFR3 and FGFR4. Inhibiting βKlotho was more effective than inhibiting FGFR4.Screening of patients who could benefit from anti-FGFR therapies.
Prostate cancer[27]Frozen primary tumor tissue, FFPE metastases. PC3, DU145, VCaP, LnCaP cancer cell lines, PNT1a cellsKLB gene expression observed with qRT-PCR in DU145 and VCaP only, and FGF19 showed stimulatory effects. βKlotho was detected in a majority of primary and metastatic lesions using IHC.βKlotho IHC may be useful for screening patients who could benefit from anti-FGFR therapy.
Breast cancer[21]Frozen tumor and adjacent tissue. MDA-MB-231 and HS578T cancer cell linesβKlotho was downregulated in cancer versus normal tissues and was undetectable in both cell lines, suggesting a tumor-suppressor role.
Pancreatic
adenocarcinoma
[44]Gene expression data from the Gene Expression Omnibus databaseHigh KLB mRNA expression is associated with increased overall survival.KLB gene expression may be useful in predicting patient survival.
Table 4. Studies Dealing with γKlotho.
Table 4. Studies Dealing with γKlotho.
Cancer TypeReferenceType of SampleMain FindingsPotential Applications
Breast cancer[21]Frozen tumor and adjacent tissue. MDA-MB-231 and HS578T cancer cell lines.LCTL gene expression is upregulated in cancer versus normal tissues, especially in triple-negative lesions, using qRT-PCR, correlating with increased cell proliferation, histological grade, TNM stage, and reduced progression-free survival. LCTL gene expression using qRT-PCR may be useful in predicting patient survival.
Prostate[47]FFPE tumor tissue from castration-resistant prostate cancer and cell lines.Higher γKlotho expression observed by IHC in tumor tissue correlates with reduced overall survival and poor response to docetaxel in patients and in a mouse xenograft model. γKlotho IHC may predict overall survival and response to docetaxel in castration-resistant prostate cancer.
Bladder cancer[22]FFPE pre-treatment tumor tissue. UMUC3, MGH-U3 and J82 cells. Higher γKlotho expression observed by IHC in muscle-invasive versus non-muscle-invasive lesions. In non-muscle-invasive lesions, γKlotho levels correlated with poor progression-free survival.γKlotho IHC may predict overall survival in patients with non-muscle-invasive bladder cancer.
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

Mota, J.; Lima, A.M.M.; Gomes, J.I.S.; Souza de Andrade, M.; Brito, H.O.; Silva, M.M.A.L.; Faustino-Rocha, A.I.; Oliveira, P.A.; Lopes, F.F.; Gil da Costa, R.M. Klotho in Cancer: Potential Diagnostic and Prognostic Applications. Diagnostics 2023, 13, 3357. https://doi.org/10.3390/diagnostics13213357

AMA Style

Mota J, Lima AMM, Gomes JIS, Souza de Andrade M, Brito HO, Silva MMAL, Faustino-Rocha AI, Oliveira PA, Lopes FF, Gil da Costa RM. Klotho in Cancer: Potential Diagnostic and Prognostic Applications. Diagnostics. 2023; 13(21):3357. https://doi.org/10.3390/diagnostics13213357

Chicago/Turabian Style

Mota, Jucileide, Alice Marques Moreira Lima, Jhessica I. S. Gomes, Marcelo Souza de Andrade, Haissa O. Brito, Melaine M. A. Lawall Silva, Ana I. Faustino-Rocha, Paula A. Oliveira, Fernanda F. Lopes, and Rui M. Gil da Costa. 2023. "Klotho in Cancer: Potential Diagnostic and Prognostic Applications" Diagnostics 13, no. 21: 3357. https://doi.org/10.3390/diagnostics13213357

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