Next Article in Journal
Genetic and Clinical Characterization of Danish Achromatopsia Patients
Previous Article in Journal
Treatment-Resistant Schizophrenia, Clozapine Resistance, Genetic Associations, and Implications for Precision Psychiatry: A Scoping Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Relevance of Telomerase and Telomere-Associated Proteins in B-Acute Lymphoblastic Leukemia

by
Tales Henrique Andrade da Mota
1,2,*,
Ricardo Camargo
3,
Estefânia Rodrigues Biojone
3,
Ana Flávia Reis Guimarães
2,
Fabio Pittella-Silva
1 and
Diêgo Madureira de Oliveira
1
1
Laboratory of Molecular Pathology of Cancer, University of Brasilia, Brasilia 70910-900, Brazil
2
Laboratory of Molecular Analysis, Faculty of Ceilândia, University of Brasilia, Brasilia 72220-275, Brazil
3
Brasília Children’s Hospital José Alencar, Brasilia 70684-831, Brazil
*
Author to whom correspondence should be addressed.
Genes 2023, 14(3), 691; https://doi.org/10.3390/genes14030691
Submission received: 10 February 2023 / Revised: 4 March 2023 / Accepted: 8 March 2023 / Published: 10 March 2023
(This article belongs to the Section Human Genomics and Genetic Diseases)

Abstract

:
Telomeres and telomerase are closely linked to uncontrolled cellular proliferation, immortalization and carcinogenesis. Telomerase has been largely studied in the context of cancer, including leukemias. Deregulation of human telomerase gene hTERT is a well-established step in leukemia development. B-acute lymphoblastic leukemia (B-ALL) recovery rates exceed 90% in children; however, the relapse rate is around 20% among treated patients, and 10% of these are still incurable. This review highlights the biological and clinical relevance of telomerase for B-ALL and the implications of its canonical and non-canonical action on signaling pathways in the context of disease and treatment. The physiological role of telomerase in lymphocytes makes the study of its biomarker potential a great challenge. Nevertheless, many works have demonstrated that high telomerase activity or hTERT expression, as well as short telomeres, correlate with poor prognosis in B-ALL. Telomerase and related proteins have been proven to be promising pharmacological targets. Likewise, combined therapy with telomerase inhibitors may turn out to be an alternative strategy for B-ALL.

Graphical Abstract

1. Introduction

Leukemia is characterized by the production of abnormal leukocytes based on cytogenetic alterations, molecular modifications, clinical features and, notably, high proliferation [1]. Although knowledge on molecular alterations that lead to leukemogenesis as well as in the mechanisms involved in disease maintenance and propagation has gradually increased over the years, the identification of novel strategies to treat the disease is still lacking. Telomeres and telomerase are closely associated with cell proliferation, which makes them attractive targets for studies in oncology [2]. Telomerase is a reverse transcriptase that elongates the telomeres, thereby compensating the loss of telomere repeats after successive replication cycles; this is a phenomenon integrated to carcinogenesis known as cell immortalization [3]. Therefore, telomerase activity (TA) is detectable in almost all types of malignant cells, including leukemia cells. In this article, we discuss the relevance of telomerase and other telomere-associated factors to B-acute lymphoblastic leukemia (B-ALL) as well as their implications for development of new treatments, highlighting the role of these proteins as potential markers of B-ALL.

2. Materials and Methods

We conducted a literature search using the NCBI database (PubMed) in September 2022 using the following combinations of keywords: (“telomerase” OR “hTERT”) AND (“Acute lymphoblastic leukemia” OR “B-ALL” OR “ALL-B” OR “ALL”) and (“telomerase” OR “hTERT”) AND (“Acute lymphoblastic leukemia”) AND “treatment”. Additional references were obtained from cross-referencing bibliographies.

3. Discussion

3.1. Acute Lymphoblastic Leukemia

Acute leukemias are characterized by an uncontrolled proliferation of myeloid precursor cells or lymphoid precursor cells, with high rates of blasts in the blood as well as predominance of malignant cells [4]. Acute lymphoblastic leukemia (ALL) originates from B-cell precursor lineages (B-ALL) or, at a lower frequency, T-cell precursor lineages (T-ALL) [5]. Both share multiple subtypes of structural chromosomal alterations—aneuploidy, chromosomal rearrangements, and mutations—which are usually related to the development of B and T cells and cell cycle regulation [6].
The ALL is responsible for over 70% of the different types of leukemia that affect children [7]. The overall 5-year event-free survival rate for this disease exceeds 90% in developed countries [8]; however, 10–20% of patients succumb to recurrences, with high lethality [9]. Unlike pediatric ALL, adult ALL historically has a poor prognosis, with limited treatment options and a cure rate under 40% [10]. Adult patients have more cooperative mutations, which promotes epigenetic modifications that can lead to B-cell development [11]. The standard treatment for acute leukemia, both in adults and children, has been focused on high-intensity induction chemotherapy. In some cases, when chemotherapy is ineffective, hematopoietic cells can be transplanted to eradicate residual disease [12]. Nevertheless, cell transplantation is not recommended for all patients [13,14,15,16]. Despite treatments having increased survival rates in most cases, it is not uncommon for patients to develop resistance to treatment or to relapse over the years [17,18], making a constant search for new therapeutic strategies necessary.
The B-ALL cells and T-ALL cells have unique molecular markers. Some of them are based on chromosomal rearrangements and lymphocyte mutations, and others, such as cytokine receptors and protein kinases, are based on the dysregulation of signaling pathways [19]. These markers make up a cellular profile that is usually associated with prognostic or response to therapy. Nevertheless, the identification of new features is beneficial to improve the predictive potential of these molecular signatures.

3.2. B-ALL and Its Molecular Markers

B-ALL is the most common cancer in children [20]. According to the 2016 classification by the World Health Organization, there are more than 11 different B-ALL types. Among them, there are the old classifications BCR-ABL1 +ALL (Ph+), BCR-ABL1-like B-ALL (Ph-like or Ph-), and the KMT2A rearrangement, also known as MLL (mixed-lineage or myeloid-lymphoid leukemia), consisting of the rearrangement of the lysine methyltransferase 2A encoding gene to a highly diverse range of partner genes [21].
The Ph+ translocation t (9;22) (q34; q11) leads to a smaller chromosome [22]. It is less present in children, but its prevalence increases with aging, resulting in a greater occurrence in adults [23]. This translocation results in expression of the fusion gene BCR-ABL1 [24]. BCR-ABL encodes a tyrosine kinase protein that promotes uncontrolled proliferation and inhibition of apoptosis [25,26]. Ph+ clinical protocols have the main focus on multiagent chemotherapy in combination with tyrosine kinase inhibitors; this combination significantly improved outcomes in adults with newly diagnosed BCR/ABL1-positive ALL [27]. However, eventual relapses are inevitable [6].
The Ph-like type affects mainly male patients, and receives its name due to its gene expression profile, which is similar to Ph+, without the BCR-ABL1 fusion protein. The Ph-like ALL patients are up to 20% adolescents and young adults, but this disease can also be diagnosed during childhood; this occurs in approximately 3%, and is associated with worse outcomes [28,29]. This type of leukemia also shows complex genomic and genetic changes that deregulate cytokine receptors and tyrosine kinase proteins [30,31,32]. Around 50% of Ph-like cases exhibit overexpression of cytokine receptor-like factor 2 (CRLF2). However, mutations in other genes such as IKZF1, ABL1, JAK2, ABL2, PDGFRB, TYK2, CSF1R, CRLF, and EPOR may also be related to this modality of B-ALL [33]. The major modifications are the translocation of the JAK-STAT family and alterations in CRLF2 [34,35,36].
The translocation t (4;11) (q21; q23) results in a genetic fusion between KMT2A-AF4, which is an alteration that mostly affects children with ALL. In addition, there are other known rearrangements involving KMT2A, such as KMT2A-AF10, KMT2A-AF9, and KMT2A-ENL [37,38,39]. These chromosomal rearrangements produce oncofusion proteins that harm the differentiation of hematopoietic stem cells [40]. The KMT2A-AFF1 translocations arise in utero and rapidly lead to the development of overt ALL. KMT2A-AF4 and AML1/MTG8 are associated with poor differentiation. This rearrangement is an important factor that supports hTERT expression due to a link between self-renewal and the transcriptional programs of leukemia cells [41].
In addition, a large number of new translocations serving as biomarkers in B-ALL have been identified, including rearrangements in DUX4, ZNF384, MEF2D, MYC and NUTM1. Mutation markers, such as PAX5-P80R and IKZF1-N159Y, are also used as biomarkers. Moreover, small telomere and telomerase activities have been reported as potential biomarkers in different oncological patients [42,43,44]. Furthermore, telomere maintenance and hTERT expression are being considered as potential targets in leukemia [45,46,47]. In this context, the relevance of using biomarkers to understand B-acute lymphoblastic leukemia, as well as for guiding therapies, is evident. The search for new markers is therefore essential to improve clinical conduct.

3.3. Telomeres, Shelterin Complex and Blood Cells

The telomeric tandem repeat sequences of TTAGGG are located in the end of chromosomes. They are bound by a specialized protein complex known as shelterin [48]. The telomeres play vital roles in cellular processes due to their capacity to protect chromosomes from end-to-end fusions and genome instability [49,50]. Cells with absent telomere maintenance mechanisms exhibit a maximum cell division capacity. Due to the loss of chromosome-capping function in telomeres, the cell enters senescence or is lead to apoptosis [51,52]. Furthermore, G-rich telomere repeat sequences are susceptible to oxidative damage, which reinforces telomere shortening and leads to cell senescence related to aging [53].
The shelterin complex is composed of six protein subunits—TRF1, TRF2, RAP1, TIN2, ACD, and POT1 (Figure 1) [54]. Although shelterin have many functions, such as protecting the telomeres from DNA deterioration and preventing activation of unwanted repair systems, they also play a key role in telomerase activity regulation [55,56,57].
Shelterin are also involved in the establishment of heterochromatin and telomeric silencing. The recruitment of these proteins is apparently related to enriching methyltransferases into the sub-telomere regions for gene silencing, which allows telomere lengthening [58].
The composition of white blood cells depends on different exposures to stress factors [59]. Different stressors can initiate a redistribution of leukocytes from immune reservoirs to the circulation. This is relevant due the fact that telomere length (TL) differs among leukocyte subtypes (lymphocytes, monocytes, granulocytes). Moreover, naïve leukocytes have telomeres similar to those found in hematopoietic stem cell progenitors, while smaller telomeres are present in mature leukocytes. This makes it difficult to define if alterations in TL can be attributed to a blood sample leukocyte composition or to a particular condition [60,61].

3.4. Telomerase and Cancer

The telomerase consists of the catalytic telomerase reverse transcriptase subunit, known as TERT, and an RNA component (hTR) that works as a template for telomere extension [62,63,64]. The canonical functions of telomerase are related to telomere length maintenance and genome stability, while the non-canonical functions are involved in the regulation of non-telomeric DNA, alterations in cell cycle kinetics, the rise of proliferation, chromatin remodeling and more (Table S1) [65,66,67].
Telomerase acts on TL maintenance during the fetal phase of life, and its presence in adult tissues is infrequent. In leukocytes, TL is stabilized around age 20, and a slow rate of attrition occurs during adulthood [68]. Moreover, strong telomerase activity is found in progenitor stem cells and activated lymphocytes, and it is especially enhanced in carcinogenesis, with implications for genome integrity, proliferation and stemness [69].
Telomerase is present in tumor cells from over 85% of cancer types, while about 15% of them continue the telomere lengthening through homologous recombination processes collectively known as alternative lengthening of telomeres (ALT), which is not a telomerase-dependent mechanism [70,71,72].
The regulation of telomerase activity is crucial and occurs mainly through the control of hTERT transcription, which also determines in which type of cell telomerase will be expressed [62]. The regulation of the active enzyme, on the other hand, is performed by a post-transcriptional maturation process involving binding to hTR (which is constantly expressed) [48]. Moreover, the regions containing TERT and hTR genes, 5p15.33 and 3q26.3, respectively, are usually amplified in cancer cells [73]. However, the hTERT mutation itself has been shown to be insufficient for telomere maintenance [74].
Mutations in the hTERT promoter represent frequent somatic genetic alterations that cause telomerase reactivation [75,76]. Epigenetic changes are also involved in different steps of this reactivation, including DNA methylation of hTERT controllers that are associated with transcription activators such as c-myc, MZF-2, and WT-1. Hypermethylation prevents binding of the repressors to the promoter, which leads to hTERT upregulation and telomerase activation [77,78].
Telomerase was first described for its capacity to elongate telomeres [79]. Nevertheless, it is becoming clear that TERT is also involved in distinct biological pathways [80] that are related to both physiological and pathological processes. These processes include those that contain stem cell functions, homeostasis, aging, tumor progression, drug resistance, regulation of non-telomeric DNA damage responses, promotion of cell growth and proliferation, acceleration of the cell cycle and damage to mitochondrial DNA, which influences cell integrity following oxidative stress. For these non-canonical activities, telomerase was reported to act on the activation of the senescence signaling pathway, the induction of apoptosis through mitochondrial pathways, autophagy, cellular growth, NF-kB mediated inflammation, and cancer progression in general (Table S1).
Telomerase expression and activity are also influenced by factors from distinct pathways. P23, for example, acts as an anti-apoptotic factor that plays a significant role in estrogen receptor α signal transduction, but which can also regulate TA by binding directly to the catalytic subunit of telomerase. This interaction is required for TL maintenance for an efficient telomerase assembly, helping to modulate telomerase–DNA binding in extension activities. Thus, the overexpression of p23 causes B-ALL cells to evade apoptosis for both TERT-related and independent pathways [81]. Similarly, c-MYC promotes hTERT deregulation, resulting in the reduction of telomere length, telomerase activity and cell proliferation [82]. Additionally, TERT can act as a transcription co-factor that regulates expression of several genes [77], which are summarized in Supplementary Table S1 [83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165].
Interestingly, there is evidence that TA is also related to gender. Male Egyptian B-ALL patients, for example, were reported to have higher expression and activity of TERT than female patients. In this particular study, the total leukocyte count of both groups of patients was higher when telomerase is upregulated, indicating a poor response to therapy [166].
The multi-functional profile of telomerase, as well as its relevance for carcinogenesis and cancer maintenance, make it an extremely relevant target for the development of studies focusing on both therapeutical purposes and on the understanding of tumor biology, especially in blood-related diseases [167,168,169].

3.5. Telomerses and Telomerase in B-Acute lymphoblastic leukemia

The hTERT mRNA can be detected in memory and naïve germinal center B-cells (GC) in which its level is associated with high TA [170,171]. The expression of telomerase in GC B-cells is inducible during immunological response, but at lower levels than in leukemic cells [172]. The synergistic stimulation with anti-IgM Ab plus specific cytokines (IL-2, IL-4, and IL-13), as well as the surface molecules BCR or CD40, increase telomerase activity in B-cells. Then, the canonical telomerase functions seem to be the main mechanism for telomere length maintenance in the germinal center in normal (non-pathological) conditions (Figure 2).
Despite the fact that up-regulation of telomerase in human B lymphocytes may occur independently of cellular proliferation, with expression of telomerase catalytic subunits [173], it has been demonstrated that telomerase activity can also be induced by PI-3 kinase-dependent and independent pathways linked to proliferation. For instance, the inhibition of PI3K blocked the anti-IgM plus anti-CD40-induced telomerase expression in B cells in a dose-dependent manner [172,174].
Telomerase is virtually absent in most adult tissues and detectable in most tumors, but the physiological role of this enzyme in lymphocytes represents an important challenge for approaching it as biological marker in leukemia. However, the straight relationship between telomerase activity and proliferation, as well as its anti-apoptotic role [175], make it essential for leukemogenesis. The uncontrolled proliferation of B lymphoblastic precursor cells in B-ALL leads to shortened telomeres and raised telomerase activity [176,177]. Additionally, leukemia cells with a normal karyotype exhibit longer telomeres when compared with cells with abnormal karyotypes [176,177].
Both high telomerase activity and shortened telomeres are correlated with disease progression, resistance to therapy and bad prognosis in ALL [178]. Telomerase can block apoptosis mechanisms in leukemic blasts, resulting in faster disease progression, and its activities are related with lactate dehydrogenase, which is an unfavorable prognostic factor for ALL patients [166]. In this sense, recent studies have revealed telomerase overexpression and hTERT methylation status as a promising prognostic biomarkers in B-ALL (especially for childhood disease), and more precisely for maintenance and disease persistence, also reinforcing the potential of telomerase as therapeutical target, mainly due to its multiple non-canonical actions [45,47,179].
In Ph+ B-ALL, the p16INK4A/pRb pathway with a high TA determines a group of adult ALL associated with poor prognosis [180]. Furthermore, Philadelphia chromosome genes also regulate telomerase and its activity at multiple levels [181]. Antisense Inhibition of BCR/ABL, for example, is able to enhance telomerase activity, leading to activation of tyrosine kinase proteins and inhibition of apoptosis [25,26].
However, controversial results can be found in the literature. In the work of Ozgur et al. [182] and Eskandari et al. [179], no significant association was found between hTERT mRNA expression and hematological parameters in B-ALL. Nevertheless, these same studies have showed that telomere attrition is linked to childhood ALL. On the other hand, Borssén et al. have demonstrated that B-cell precursor group cases had a higher hTERT methylation than diploid ALL. In addition, hTERT mRNA levels were negatively associated with methylation status, but curiously, in low-risk B-cell precursor patients, long telomeres indicated a worse prognosis [183].
Monitoring minimal residual disease (MRD) is one of the most important strategies to follow up B-ALL patients due the capacity to identify lower cell levels. In this sense, it was demonstrated that quantification of telomerase expression along with monitoring MRD by qPCR can strengthen the follow up of patients with B-ALL. This would not only improve treatment follow up but also help to identify post-therapy remission [47].
Finally, a large number of studies propose that pathogenesis and the phenotypic characteristics of B-acute lymphoblastic leukemia are connected with the conjunction of specific targets and DNA variations promoted by epigenetic alterations such as methylation [184,185,186]. hTERT promoter methylation is infrequent in B-ALL cases with remission, and there is no association with TL. However, hTERT RNA expression is reduced when methylation occurs [183]. Methylation of CDKN2B CpG island was associated with high telomerase activity in children with B-ALL [187]. It has also been shown that β-Arrestin1 promotes cellular senescence in B-ALL by binding with P300-Sp1 in order to regulate hTERT transcription. In that case, hTERT is a major factor due to the regulation stimulated on the β-Arrestin pathway, rising p300-sp1 expression [87].

3.6. Shelterin in B Lymphoblastic Leukemia

The role of the shelterin complex in B-ALL has also been studied. TRF2 expression was shown to be increasing in acute leukemias and also higher in lymphocytes of B-ALL patients, particularly in those with an abnormal karyotype [177]. Recently, NOTCH3, PAX5, CBFB, and particularly ACD were shown to drive the activated RAS pathway and monosomy 7 to B-acute lymphoblastic leukemia [188]. Nonetheless, ACD plays a key role in telomere maintenance due to its interaction with POT1; this combination protects telomeres and recruit telomerase at chromosome ends. Despite the overexpression of wild-type, ACD does not lead to telomere lengthening, the G223V mutation reflects on TL and seems to be related to decreased apoptosis activity in B-ALL cells, that is triggered to the functional role of ACD and its relevance for cell survival in leukemia [189].
Beyond hTERT, B-ALL patients also show high expression of CTC1 and OBFC1 (they are part of CST complex which works with the shelterin complex to lengthen telomeres); however, only CTC1 was associated with leukemia [190].

3.7. Telomerase and Genetic Variation

There is, currently, a vast field literature on hTERT polymorphisms and their implications in oncology, but just few works approach it in the context of B-ALL. The hTERT polymorphisms rs2735940 and rs2736100, for example, were defined as risk factor for ALL and turned out to be functional; they were implicated in TA, TL and homeostasis. The same authors showed that a variant near hTR, as well as high TL, are markers for risk of acute lymphoblastic leukemia in Chinese children [191].
Another study demonstrated that the survival rate of children with B-ALL was higher in European American children (EA) than in African American children (AA), which appeared to be due to the different canonical pathways affected in each case. Telomerase signaling is related to AA pathways, while chromosome aberrations in EA more frequently affect genes involved with homologous recombination [192]. This suggests that hTERT may have a different influence on B-ALL with regard to different populations; nonetheless, large-scale studies need to be done to verify this hypothesis.

3.8. Current Telomerase Inhibitors and Their Clinical Potential

Different approaches for telomerase inhibition have been under development for more than a decade, aiming at more effective treatment strategies. Telomerase activity can be inhibited by different strategies, such as disrupting biosynthesis, maturation, assembly, or correct interaction between the telomerase complex and the substrate [193]. In Table 1, we exemplified some of the currently available telomerase inhibitors.
Doxorubicin (DOX) is a chemotherapy drug used in different cancer treatment protocols. This molecule promotes cell death through disruption of DNA repair by inhibiting topoisomerase II, and provokes oxidative stress by generating free radicals [7,206]. However, doxorubicin has a high toxicity for the heart, which can lead to mortality among cancer patients, limiting its clinical applications [207].
The Zataria multiflora extract (ZME) is a plant extract oil that exhibited a synergistic effect in association with doxorubicin, increasing its toxicity in all tested B-ALL cell lines. Despite this combination raising the levels of anti-apoptotic Bcl-2, it downregulated expression of c-Myc and hTERT, showing ZME as a potential adjuvant for treatment of pre-B-acute lymphoblastic leukemia [198]. Additionally, the telomerase inhibitor MST-312 decreased in vitro effective dose of doxorubicin. The combination MST-312/DOX reduced cell growth and promoted apoptosis in -B-ALL cells through unbalancing the Bax/Bcl-2 ratio aligned to down-regulation of c-Myc and hTERT [208]. It is important to mention that most TERT inhibitors are developed, aiming canonical function of telomerase, but there is evidence of the antitumor effect of MST-312 associated with non-canonical ones [207].
BIBR1532 is one of the most powerful telomerase inhibitors. This synthetic non-nucleoside compound binds to telomerase and acts as a chain terminator during nucleotide polymerization, inhibiting TA in a dose-dependent way [209,210]. BIBR1532 provoked cell death in pre-B-ALL cells after suppression of hTERT and c-Myc expression. Besides, high doses of BIBR1532 can induce p73, up-regulate Bax and activate caspase-3 [46]. Association of BIBR1532 with doxorubicin also reduced surviving expression and produced a synergistic anticancer effect in B-ALL through induction of ROS, which increased expression of Bax. Furthermore, it raised p21 levels, which promoted G1 cell cycle arrest and downregulation of p73-mediated c-Myc and hTERT expression [211].
To summarise, the combination of DOX with BIBR1532, ZME or MST-312 increases its therapeutic effect. A synergistic mechanism, if confirmed, could lead to therapeutical protocols with a lower dose of doxorubicin, thereby decreasing the risk of DOX-induced cardiotoxicity.
It is important to mention that there are a vast number of works showing the antitumor effects of telomerase inhibitors in a variety of in vitro and in vivo models of different cancer types [212], including some with which clinical trials are in progress or already concluded [213]. There are also studies testing telomerase-targeted immunotherapy [214] and other telomere-related therapeutical strategies. In this review, we attempted to summarize information regarding telomerase in B-ALL, which is still very scarce. Nevertheless, any one of these prototypes, once proved effective for cancer control, has the potential to be used in the context of leukemia.
In any case, further investigation is deeply required, including clinical trials, which could determine the safety of these compounds alone or as combined therapies for B-ALL patients. However, the greatest challenge that must be overcome in order to take studies with telomerase inhibition to the next level is the development of compounds targeting inhibition of non-canonical functions, which have been demonstrated to be crucial for cancer maintenance.

4. Conclusions

The physiological functions of telomerase in lymphocytes represent a challenge to determine the role of this enzyme in B-ALL. However, clearly reviewed data showed evidence of the potential of telomerase and other telomere-related proteins as clinical biomarkers and pharmacological targets. Briefly, high telomerase activity or hTERT expression, as well as short lymphocytes’ telomeres, are frequently correlated with poor prognosis or even higher risk for B-ALL. However, there are still some apparent conflicting data in the literature, associating long telomeres with worse prognosis. We emphasize the word “apparent” since it also became clear that there is no single pattern concerning telomere and telomerase functions in leukemia, especially considering all possibilities of non-canonical actions of TERT. Additionally, the influence of telomerase on B-ALL seems to be divergent in different ethnic groups, which needs further investigation to be better elucidated. Finally, this pool of results shows a promising future for telomere and telomerase targeted therapy as new or combined treatments, but most data are too preliminary for short-term clinical use, especially in ALL.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/genes14030691/s1, Table S1: Markers associated to canonical and non-canonical functions of telomerase.

Author Contributions

T.H.A.d.M.: Writing—original draft; R.C.: Supervision; E.R.B.: Writing—review and editing; A.F.R.G.: Writing—original draft; F.P.-S.: Writing—review and editing; D.M.d.O.: Conceptualization, Supervision. All authors have read and agreed to the published version of the manuscript.

Funding

The authors receive fundings from the Brazilian Council for Scientific and Technological Development (CNPq), grant number 88887.501384/2020-00, and from the Fundação de Apoio à Pesquisa do Distrito Federal (FAPDF), grant numbers 00193-00001029/2021-95 and 00193-00000891/2021-81.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors would like to acknowledge the Brazilian Council for Scientific and Technological Development (CNPq) and the Fundação de Apoio à Pesquisa do Distrito Federal (FAPDF), for their support.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Whiteley, A.E.; Price, T.T.; Cantelli, G.; Sipkins, D.A. Leukaemia: A Model Metastatic Disease. Nat. Rev. Cancer 2021, 21, 461–475. [Google Scholar] [CrossRef]
  2. Lin, J.; Epel, E. Stress and Telomere Shortening: Insights from Cellular Mechanisms. Ageing Res. Rev. 2022, 73, 101507. [Google Scholar] [CrossRef] [PubMed]
  3. Srinivas, N.; Rachakonda, S.; Kumar, R. Telomeres and Telomere Length: A General Overview. Cancers 2020, 12, 558. [Google Scholar] [CrossRef] [Green Version]
  4. Roussel, X.; Daguindau, E.; Berceanu, A.; Desbrosses, Y.; Warda, W.; Neto da Rocha, M.; Trad, R.; Deconinck, E.; Deschamps, M.; Ferrand, C. Acute Myeloid Leukemia: From Biology to Clinical Practices Through Development and Pre-Clinical Therapeutics. Front. Oncol. 2020, 10, 599933. [Google Scholar] [CrossRef] [PubMed]
  5. Tran, T.H.; Hunger, S.P. The Genomic Landscape of Pediatric Acute Lymphoblastic Leukemia and Precision Medicine Opportunities. Semin. Cancer Biol. 2022, 84, 144–152. [Google Scholar] [CrossRef]
  6. Inaba, H.; Mullighan, C.G. Pediatric Acute Lymphoblastic Leukemia. Haematologica 2020, 105, 2524–2539. [Google Scholar] [CrossRef] [PubMed]
  7. Aberuyi, N.; Rahgozar, S.; Ghodousi, E.S.; Ghaedi, K. Drug Resistance Biomarkers and Their Clinical Applications in Childhood Acute Lymphoblastic Leukemia. Front. Oncol. 2020, 9, 1496. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Kaplan, J.A. Leukemia in Children. Pediatr. Rev. 2019, 40, 319–331. [Google Scholar] [CrossRef]
  9. Waanders, E.; Gu, Z.; Dobson, S.M.; Antić, Ž.; Crawford, J.C.; Ma, X.; Edmonson, M.N.; Payne-Turner, D.; van de Vorst, M.; Jongmans, M.C.J.; et al. Mutational Landscape and Patterns of Clonal Evolution in Relapsed Pediatric Acute Lymphoblastic Leukemia. Blood Cancer Discov. 2020, 1, 96–111. [Google Scholar] [CrossRef]
  10. Samra, B.; Jabbour, E.; Ravandi, F.; Kantarjian, H.; Short, N.J. Evolving Therapy of Adult Acute Lymphoblastic Leukemia: State-of-the-Art Treatment and Future Directions. J. Hematol. Oncol. 2020, 13, 70. [Google Scholar] [CrossRef]
  11. Liu, Y.-F.; Wang, B.-Y.; Zhang, W.-N.; Huang, J.-Y.; Li, B.-S.; Zhang, M.; Jiang, L.; Li, J.-F.; Wang, M.-J.; Dai, Y.-J.; et al. Genomic Profiling of Adult and Pediatric B-Cell Acute Lymphoblastic Leukemia. EBioMedicine 2016, 8, 173–183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Malczewska, M.; Kośmider, K.; Bednarz, K.; Ostapińska, K.; Lejman, M.; Zawitkowska, J. Recent Advances in Treatment Options for Childhood Acute Lymphoblastic Leukemia. Cancers 2022, 14, 2021. [Google Scholar] [CrossRef] [PubMed]
  13. Li, H.; Yang, Y.; Hong, W.; Huang, M.; Wu, M.; Zhao, X. Applications of Genome Editing Technology in the Targeted Therapy of Human Diseases: Mechanisms, Advances and Prospects. Signal Transduct. Target. Ther. 2020, 5, 1. [Google Scholar] [CrossRef] [Green Version]
  14. Kato, M.; Manabe, A. Treatment and Biology of Pediatric Acute Lymphoblastic Leukemia. Pediatr. Int. 2018, 60, 4–12. [Google Scholar] [CrossRef] [PubMed]
  15. Wu, Y.W.; Chao, M.W.; Tu, H.J.; Chen, L.C.; Hsu, K.C.; Liou, J.P.; Yang, C.R.; Yen, S.C.; HuangFu, W.C.; Pan, S.L. A Novel Dual HDAC and HSP90 Inhibitor, MPT0G449, Downregulates Oncogenic Pathways in Human Acute Leukemia in Vitro and in Vivo. Oncogenesis 2021, 10, 39. [Google Scholar] [CrossRef] [PubMed]
  16. Boyiadzis, M.M.; Aksentijevich, I.; Arber, D.A.; Barrett, J.; Brentjens, R.J.; Brufsky, J.; Cortes, J.; De Lima, M.; Forman, S.J.; Fuchs, E.J.; et al. The Society for Immunotherapy of Cancer (SITC) Clinical Practice Guideline on Immunotherapy for the Treatment of Acute Leukemia. J. Immunother. Cancer 2020, 8, e001235. [Google Scholar] [CrossRef]
  17. Autry, R.J.; Paugh, S.W.; Carter, R.; Shi, L.; Liu, J.; Daniel, C.; Lau, C.E.; Bonten, E.J.; Yang, W.; Mccorkle, J.R.; et al. Integrative Fenomic Analyses Reveal Mechanisms of Glucocorticoid Resistane in A. Nat. Cancer 2020, 1, 329–344. [Google Scholar] [CrossRef]
  18. Su, Q.; Fan, Z.; Huang, F.; Xu, N.; Nie, D.; Lin, D.; Guo, Z.; Shi, P.; Wang, Z.; Jiang, L.; et al. Comparison of Two Strategies for Prophylactic Donor Lymphocyte Infusion in Patients With Refractory/Relapsed Acute Leukemia. Front. Oncol. 2021, 11, 554503. [Google Scholar] [CrossRef]
  19. Iacobucci, I.; Mullighan, C.G. Genetic Basis of Acute Lymphoblastic Leukemia. J. Clin. Oncol. 2017, 35, 975–983. [Google Scholar] [CrossRef]
  20. Maamari, D.; El-Khoury, H.; Saifi, O.; Muwakkit, S.A.; Zgheib, N.K. Implementation of Pharmacogenetics to Individualize Treatment Regimens for Children with Acute Lymphoblastic Leukemia. Pharmgenomic. Pers. Med. 2020, 13, 295–317. [Google Scholar] [CrossRef] [PubMed]
  21. Rabin, K.R. Genetic Ancestry and Childhood Acute Lymphoblastic Leukemia Subtypes and Outcomes in the Genomic Era. JAMA Oncol. 2022, 8, 342. [Google Scholar] [CrossRef]
  22. Komorowski, L.; Fidyt, K.; Patkowska, E.; Firczuk, M. Philadelphia Chromosome-Positive Leukemia in the Lymphoid Lineage—Similarities and Differences with the Myeloid Lineage and Specific Vulnerabilities. Int. J. Mol. Sci. 2020, 21, 5776. [Google Scholar] [CrossRef] [PubMed]
  23. Xiao, X.; Xiao, X.; Liu, P.; Liu, P.; Li, D.; Li, D.; Xia, Z.; Wang, P.; Wang, P.; Zhang, X.; et al. Combination Therapy of BCR-ABL-Positive B Cell Acute Lymphoblastic Leukemia by Tyrosine Kinase Inhibitor Dasatinib and c-JUN N-Terminal Kinase Inhibition. J. Hematol. Oncol. 2020, 13, 80. [Google Scholar] [CrossRef]
  24. Brown, L.M.; Hediyeh-zadeh, S.; Sadras, T.; Huckstep, H.; Sandow, J.J.; Bartolo, R.C.; Kosasih, H.J.; Davidson, N.M.; Schmidt, B.; Bjelosevic, S.; et al. SFPQ-ABL1 and BCR-ABL1 Use Different Signaling Networks to Drive B-Cell Acute Lymphoblastic Leukemia. Blood Adv. 2022, 6, 2373–2387. [Google Scholar] [CrossRef]
  25. Bakalova, R.; Ohba, H.; Zhelev, Z.; Kubo, T.; Fujii, M.; Ishikawa, M.; Shinohara, Y.; Baba, Y. Antisense Inhibition of Bcr-Abl/c-Abl Synthesis Promotes Telomerase Activity and Upregulates Tankyrase in Human Leukemia Cells. FEBS Lett. 2004, 564, 73–84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Zhang, P.; Wang, Y.; Qin, M.; Li, D.; Odhiambo, W.O.; Yuan, M.; Lv, Z.; Liu, C.; Ma, Y.; Dong, Y.; et al. Involvement of Blnk and Foxo1 in Tumor Suppression in BCR-ABL1-Transformed pro-B Cells. Oncol. Rep. 2021, 45, 693–705. [Google Scholar] [CrossRef]
  27. Inaba, H.; Pui, C.-H. Advances in the Diagnosis and Treatment of Pediatric Acute Lymphoblastic Leukemia. J. Clin. Med. 2021, 10, 1926. [Google Scholar] [CrossRef]
  28. Tasian, S.K.; Loh, M.L.; Hunger, S.P. Philadelphia Chromosome–like Acute Lymphoblastic Leukemia. Blood 2017, 130, 2064–2072. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Chen, Y.; Lu, G.P. Advances in the Diagnosis and Treatment of Pediatric Acute Respiratory Distress Syndrome. Chin. J. Contemp. Pediatr. 2018, 20, 717–723. [Google Scholar] [CrossRef]
  30. Bei, Y. Targeted Therapy or Transplantation for Paediatric ABL-Class Ph-like Acute Lymphocytic Leukaemia? Physiol. Behav. 2020, 7, e858–e859. [Google Scholar] [CrossRef]
  31. Hurtz, C.; Wertheim, G.B.; Loftus, J.P.; Blumenthal, D.; Lehman, A.; Li, Y.; Bagashev, A.; Manning, B.; Cummins, K.D.; Burkhardt, J.K.; et al. Oncogene-Independent BCR-like Signaling Adaptation Confers Drug Resistance in Ph-like ALL. J. Clin. Invest. 2020, 130, 3637–3653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Terwilliger, T.; Abdul-Hay, M. Acute Lymphoblastic Leukemia: A Comprehensive Review and 2017 Update. Blood Cancer J. 2017, 7, e577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Lejman, M.; Chałupnik, A.; Chilimoniuk, Z.; Dobosz, M. Genetic Biomarkers and Their Clinical Implications in B-Cell Acute Lymphoblastic Leukemia in Children. Int. J. Mol. Sci. 2022, 23, 2755. [Google Scholar] [CrossRef]
  34. Hsu, Y.C.; Yu, C.H.; Chen, Y.M.; Roberts, K.G.; Ni, Y.L.; Lin, K.H.; Jou, S.T.; Lu, M.Y.; Chen, S.H.; Wu, K.H.; et al. Philadelphia Chromosome-Negative B-Cell Acute Lymphoblastic Leukaemia with Kinase Fusions in Taiwan. Sci. Rep. 2021, 11, 5802. [Google Scholar] [CrossRef] [PubMed]
  35. Tosi, M.; Spinelli, O.; Leoncin, M.; Cavagna, R.; Pavoni, C.; Lussana, F.; Intermesoli, T.; Frison, L.; Perali, G.; Carobolante, F.; et al. Mrd-based Therapeutic Decisions in Genetically Defined Subsets of Adolescents and Young Adult Philadelphia-negative All. Cancers 2021, 13, 2108. [Google Scholar] [CrossRef] [PubMed]
  36. Harvey, R.C.; Tasian, S.K. Clinical Diagnostics and Treatment Strategies for Philadelphia Chromosome-like Acute Lymphoblastic Leukemia. Blood Adv. 2020, 4, 218–228. [Google Scholar] [CrossRef] [Green Version]
  37. Jenkins, T.W.; Kopyscinski, S.L.D.; Fields, J.L.; Rahme, G.J.; Colley, W.C.; Israel, M.A. Activity of Immunoproteasome Inhibitor ONX-0914 in Acute Lymphoblastic Leukemia Expressing MLL-AF4 Fusion Protein. Sci. Rep. 2021, 11, 10883. [Google Scholar] [CrossRef]
  38. Antunes, E.T.B.; Ottersbach, K. The MLL/SET Family and Haematopoiesis. Biochim. Biophys. Acta-Gene Regul. Mech. 2020, 1863, 194579. [Google Scholar] [CrossRef]
  39. Cao, L.; Mitra, P.; Gonda, T.J. The Mechanism of MYB Transcriptional Regulation by MLL-AF9 Oncoprotein. Sci. Rep. 2019, 9, 20084. [Google Scholar] [CrossRef] [Green Version]
  40. Wen, J.; Zhou, M.; Shen, Y.; Long, Y.; Guo, Y.; Song, L.; Xiao, J. Poor Treatment Responses Were Related to Poor Outcomes in Pediatric B Cell Acute Lymphoblastic Leukemia with KMT2A Rearrangements. BMC Cancer 2022, 22, 859. [Google Scholar] [CrossRef]
  41. Gessner, A.; Thomas, M.; Garrido Castro, P.; Büchler, L.; Scholz, A.; Brümmendorf, T.H.; Martinez Soria, N.; Vormoor, J.; Greil, J.; Heidenreich, O. Leukemic Fusion Genes MLL/AF4 and AML1/MTG8 Support Leukemic Self-Renewal by Controlling Expression of the Telomerase Subunit TERT. Leukemia 2010, 24, 1751–1759. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Engelhardt, M.; Ozkaynak, M.; Drullinsky, P.; Sandoval, C.; Tugal, O.; Jayabose, S.; Moore, M. Telomerase Activity and Telomere Length in Pediatric Patients with Malignancies Undergoing Chemotherapy. Leukemia 1998, 12, 13–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Da Silva, R.S.; de Moraes, L.S.; da Rocha, C.A.M.; Ferreira-Fernandes, H.; Yoshioka, F.K.N.; Rey, J.A.; Pinto, G.R.; Burbano, R.R. Telomere Length and Telomerase Activity of Leukocytes as Biomarkers of Selective Serotonin Reuptake Inhibitor Responses in Patients with Major Depressive Disorder. Psychiatr. Genet. 2022, 32, 34–36. [Google Scholar] [CrossRef] [PubMed]
  44. Afshari, N.; Al-Gazally, M.E.; Rasulova, I.; Jalil, A.T.; Matinfar, S.; Momeninejad, M. Sensitive Bioanalytical Methods for Telomerase Activity Detection: A Cancer Biomarker. Anal. Methods 2022, 14, 4174–4184. [Google Scholar] [CrossRef]
  45. Karow, A.; Haubitz, M.; Oppliger Leibundgut, E.; Helsen, I.; Preising, N.; Steiner, D.; Dantonello, T.M.; Ammann, R.A.; Roessler, J.; Kartal-Kaess, M.; et al. Targeting Telomere Biology in Acute Lymphoblastic Leukemia. Int. J. Mol. Sci. 2021, 22, 6653. [Google Scholar] [CrossRef]
  46. Bashash, D.; Ghaffari, S.H.; Mirzaee, R.; Alimoghaddam, K.; Ghavamzadeh, A. Telomerase Inhibition by Non-Nucleosidic Compound BIBR1532 Causes Rapid Cell Death in Pre-B Acute Lymphoblastic Leukemia Cells. Leuk. Lymphoma 2013, 54, 561–568. [Google Scholar] [CrossRef]
  47. Nogueira, B.M.D.; da Costa Pantoja, L.; da Silva, E.L.; Mello Júnior, F.A.R.; Teixeira, E.B.; Wanderley, A.V.; da Silva Maués, J.H.; de Moraes Filho, M.O.; de Moraes, M.E.A.; Montenegro, R.C.; et al. Telomerase (HTERT) Overexpression Reveals a Promising Prognostic Biomarker and Therapeutical Target in Different Clinical Subtypes of Pediatric Acute Lymphoblastic Leukaemia. Genes 2021, 12, 1632. [Google Scholar] [CrossRef]
  48. Caitlin, M.; Roake and Steven, E. Artandi Regulation of Human Telomerase in Homeostasis and Disease. Physiol. Behav. 2020, 21, 384–397. [Google Scholar] [CrossRef]
  49. Tomita, K. How Long Does Telomerase Extend Telomeres? Regulation of Telomerase Release and Telomere Length Homeostasis. Curr. Genet. 2018, 64, 1177–1181. [Google Scholar] [CrossRef] [Green Version]
  50. Mei, Y.; Deng, Z.; Vladimirova, O.; Gulve, N.; Johnson, F.B.; Drosopoulos, W.C.; Schildkraut, C.L.; Lieberman, P.M. TERRA G-Quadruplex RNA Interaction with TRF2 GAR Domain Is Required for Telomere Integrity. Sci. Rep. 2021, 11, 3509. [Google Scholar] [CrossRef]
  51. Sieverling, L.; Hong, C.; Koser, S.D.; Ginsbach, P.; Kleinheinz, K.; Hutter, B.; Braun, D.M.; Cortés-Ciriano, I.; Xi, R.; Kabbe, R.; et al. Genomic Footprints of Activated Telomere Maintenance Mechanisms in Cancer. Nat. Commun. 2020, 11, 733. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Vaiserman, A.; Krasnienkov, D. Telomere Length as a Marker of Biological Age: State-of-the-Art, Open Issues, and Future Perspectives. Front. Genet. 2021, 11, 630186. [Google Scholar] [CrossRef]
  53. Von Zglinicki, T.; Wan, T.; Miwa, S. Senescence in Post-Mitotic Cells: A Driver of Aging? Antioxid. Redox Signal. 2021, 34, 308–323. [Google Scholar] [CrossRef] [PubMed]
  54. Pan, H.; Kaur, P.; Barnes, R.; Detwiler, A.C.; Sanford, S.L.; Liu, M.; Xu, P.; Mahn, C.; Tang, Q.; Hao, P.; et al. Structure, Dynamics, and Regulation of TRF1-TIN2-Mediated Trans- And Cis-Interactions on Telomeric DNA. J. Biol. Chem. 2021, 297, 101080. [Google Scholar] [CrossRef]
  55. Erdel, F.; Kratz, K.; Willcox, S.; Griffith, J.D.; Greene, E.C.; de Lange, T. Telomere Recognition and Assembly Mechanism of Mammalian Shelterin. Cell Rep. 2017, 18, 41–53. [Google Scholar] [CrossRef] [PubMed]
  56. Kim, J.-K.; Liu, J.; Hu, X.; Yu, C.; Roskamp, K.; Sankaran, B.; Huang, L.; Komives, E.A.; Qiao, F. Structural Basis for Shelterin Bridge Assembly. Mol. Cell 2017, 68, 698–714.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Sekne, Z.; Ghanim, G.E.; van Roon, A.-M.M.; Nguyen, T.H.D. Structural Basis of Human Telomerase Recruitment by TPP1-POT1. Science 2022, 375, 1173–1176. [Google Scholar] [CrossRef]
  58. Liu, J.; Hu, X.; Bao, K.; Kim, J.K.; Zhang, C.; Jia, S.; Qiao, F. The Cooperative Assembly of Shelterin Bridge Provides a Kinetic Gateway That Controls Telomere Length Homeostasis. Nucleic Acids Res. 2021, 49, 8110–8119. [Google Scholar] [CrossRef]
  59. Semeraro, M.D.; Smith, C.; Kaiser, M.; Levinger, I.; Duque, G.; Gruber, H.-J.; Herrmann, M. Physical Activity, a Modulator of Aging through Effects on Telomere Biology. Aging 2020, 12, 13803–13823. [Google Scholar] [CrossRef]
  60. Krasnienkov, D.S.; Khalangot, M.D.; Kravchenko, V.I.; Kovtun, V.A.; Guryanov, V.G.; Chizhova, V.P.; Korkushko, O.V.; Shatilo, V.B.; Kukharsky, V.M.; Vaiserman, A.M. Hyperglycemia Attenuates the Association between Telomere Length and Age in Ukrainian Population. Exp. Gerontol. 2018, 110, 247–252. [Google Scholar] [CrossRef]
  61. Hastings, W.J.; Shalev, I.; Belsky, D.W. Translating Measures of Biological Aging to Test Effectiveness of Geroprotective Interventions: What Can We Learn from Research on Telomeres? Front. Genet. 2017, 8, 164. [Google Scholar] [CrossRef] [Green Version]
  62. Sharma, S.; Chowdhury, S. Emerging Mechanisms of Telomerase Reactivation in Cancer. Trends Cancer 2022, 8, 632–641. [Google Scholar] [CrossRef]
  63. Jacczak, B.; Rubiś, B.; Totoń, E. Potential of Naturally Derived Compounds in Telomerase and Telomere Modulation in Skin Senescence and Aging. Int. J. Mol. Sci. 2021, 22, 6381. [Google Scholar] [CrossRef]
  64. Dogan, F.; Forsyth, N.R. Telomerase Regulation: A Role for Epigenetics. Cancers 2021, 13, 1213. [Google Scholar] [CrossRef]
  65. Udroiu, I.; Marinaccio, J.; Sgura, A. Many Functions of Telomerase Components: Certainties, Doubts, and Inconsistencies. Int. J. Mol. Sci. 2022, 23, 15189. [Google Scholar] [CrossRef]
  66. Thompson, C.A.H.; Wong, J.M.Y. Non-Canonical Functions of Telomerase Reverse Transcriptase: Emerging Roles and Biological Relevance. Curr. Top. Med. Chem. 2020, 20, 498–507. [Google Scholar] [CrossRef] [PubMed]
  67. Rosen, J.; Jakobs, P.; Ale-Agha, N.; Altschmied, J.; Haendeler, J. Non-Canonical Functions of Telomerase Reverse Transcriptase–Impact on Redox Homeostasis. Redox Biol. 2020, 34, 101543. [Google Scholar] [CrossRef]
  68. Demerath, E.W.; Cameron, N.; Gillman, M.W.; Towne, B.; Siervogel, R.M. Telomeres and Telomerase in the Fetal Origins of Cardiovascular Disease: A Review. Hum. Biol. 2004, 76, 127–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Celtikci, B.; Erkmen, G.K.; Dikmen, Z.G. Regulation and Effect of Telomerase and Telomeric Length in Stem Cells. Curr. Stem Cell Res. Ther. 2021, 16, 809–823. [Google Scholar] [CrossRef]
  70. Schaich, M.A.; Sanford, S.L.; Welfer, G.A.; Johnson, S.A.; Khoang, T.H.; Opresko, P.L.; Freudenthal, B.D. Mechanisms of Nucleotide Selection by Telomerase. eLife 2020, 9, e55438. [Google Scholar] [CrossRef] [PubMed]
  71. Radu, L.E.; Colita, A.; Pasca, S.; Tomuleasa, C.; Popa, C.; Serban, C.; Gheorghe, A.; Serbanica, A.; Jercan, C.; Marcu, A.; et al. Day 15 and Day 33 Minimal Residual Disease Assessment for Acute Lymphoblastic Leukemia Patients Treated According to the BFM ALL IC 2009 Protocol: Single-Center Experience of 133 Cases. Front. Oncol. 2020, 10, 923. [Google Scholar] [CrossRef] [PubMed]
  72. Chan, W.F.; Coughlan, H.D.; Zhou, J.H.S.; Keenan, C.R.; Bediaga, N.G.; Hodgkin, P.D.; Smyth, G.K.; Johanson, T.M.; Allan, R.S. Pre-Mitotic Genome Re-Organisation Bookends the B Cell Differentiation Process. Nat. Commun. 2021, 12, 1344. [Google Scholar] [CrossRef] [PubMed]
  73. Cao, Y.; Bryan, T.M.; Reddel, R.R. Increased Copy Number of the TERT and TERC Telomerase Subunit Genes in Cancer Cells. Cancer Sci. 2008, 99, 1092–1099. [Google Scholar] [CrossRef] [PubMed]
  74. Barthel, F.P.; Wei, W.; Tang, M.; Martinez-Ledesma, E.; Hu, X.; Amin, S.B.; Akdemir, K.C.; Seth, S.; Song, X.; Wang, Q.; et al. Systematic Analysis of Telomere Length and Somatic Alterations in 31 Cancer Types. Nat. Genet. 2017, 49, 349–357. [Google Scholar] [CrossRef]
  75. Guo, Y.; Chen, Y.; Zhang, L.; Ma, L.; Jiang, K.; Yao, G.; Zhu, L. TERT Promoter Mutations and Telomerase in Melanoma. J. Oncol. 2022, 2022, 6300329. [Google Scholar] [CrossRef]
  76. Heidenreich, B.; Kumar, R. TERT Promoter Mutations in Telomere Biology. Mutat. Res.-Rev. Mutat. Res. 2017, 771, 15–31. [Google Scholar] [CrossRef]
  77. Kyo, S.; Takakura, M.; Fujiwara, T.; Inoue, M. Understanding and Exploiting HTERT Promoter Regulation for Diagnosis and Treatment of Human Cancers. Cancer Sci. 2008, 99, 1528–1538. [Google Scholar] [CrossRef] [Green Version]
  78. Chiba, K.; Lorbeer, F.K.; Shain, A.H.; McSwiggen, D.T.; Schruf, E.; Oh, A.; Ryu, J.; Darzacq, X.; Bastian, B.C.; Hockemeyer, D. Mutations in the Promoter of the Telomerase Gene TERT Contribute to Tumorigenesis by a Two-Step Mechanism. Science 2017, 357, 1416–1420. [Google Scholar] [CrossRef] [Green Version]
  79. Li, H.; Wang, B.; Li, D.; Li, J.; Luo, Y.; Dan, J. Roles of Telomeres and Telomerase in Age-Related Renal Diseases (Review). Mol. Med. Rep. 2021, 23, 96. [Google Scholar] [CrossRef]
  80. Robinson, N.J.; Schiemann, W.P. Telomerase in Cancer: Function, Regulation, and Clinical Translation. Cancers 2022, 14, 808. [Google Scholar] [CrossRef]
  81. Liu, X.; Zou, L.; Zhu, L.; Zhang, H.; Du, C.; Li, Z.; Gao, C.; Zhao, X.; Bao, S.; Zheng, H. MiRNA Mediated Up-Regulation of Cochaperone P23 Acts as an Anti-Apoptotic Factor in Childhood Acute Lymphoblastic Leukemia. Leuk. Res. 2012, 36, 1098–1104. [Google Scholar] [CrossRef]
  82. Sheikh-Zeineddini, N.; Bashash, D.; Safaroghli-Azar, A.; Riyahi, N.; Shabestari, R.M.; Janzamin, E.; Safa, M. Suppression of C-Myc Using 10058-F4 Exerts Caspase-3-dependent Apoptosis and Intensifies the Antileukemic Effect of Vincristine in Pre-B Acute Lymphoblastic Leukemia Cells. J. Cell. Biochem. 2019, 120, 14004–14016. [Google Scholar] [CrossRef] [PubMed]
  83. Cao, Y.; Li, H.; Deb, S.; Liu, J.P. TERT Regulates Cell Survival Independent of Telomerase Enzymatic Activity. Oncogene 2002, 21, 3130–3138. [Google Scholar] [CrossRef] [Green Version]
  84. Lin, S.Y.; Elledge, S.J. Multiple Tumor Suppressor Pathways Negatively Regulate Telomerase. Cell 2003, 113, 881–889. [Google Scholar] [CrossRef] [Green Version]
  85. Zhang, Y.; Toh, L.; Lau, P.; Wang, X. Human Telomerase Reverse Transcriptase (HTERT) Is a Novel Target of the Wnt/β-Catenin Pathway in Human Cancer. J. Biol. Chem. 2012, 287, 32494–32511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Linne, H.; Yasaei, H.; Marriott, A.; Harvey, A.; Mokbel, K.; Newbold, R.; Roberts, T. Functional Role of SETD2, BAP1, PARP-3 and PBRM1 Candidate Genes on the Regulation of HTERT Gene Expression. Oncotarget 2017, 8, 61890–61900. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Liu, S.; Liu, H.; Qin, R.; Shu, Y.; Liu, Z.; Zhang, P.; Duan, C.; Hong, D.; Yu, J.; Zou, L. The Cellular Senescence of Leukemia-Initiating Cells from Acute Lymphoblastic Leukemia Is Postponed by β-Arrestin1 Binding with P300-Sp1 to Regulate HTERT Transcription. Cell Death Dis. 2017, 8, e2756. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Mandal, M.; Kumar, R. Bcl-2 Modulates Telomerase Activity. J. Biol. Chem. 1997, 272, 14183–14187. [Google Scholar] [CrossRef] [Green Version]
  89. Jin, Y.; You, L.; Kim, H.J.; Lee, H.-W. Telomerase Reverse Transcriptase Contains a BH3-Like Motif and Interacts with BCL-2 Family Members. Mol. Cells 2018, 41, 684–694. [Google Scholar] [CrossRef]
  90. Ding, X.; Nie, Z.; She, Z.; Bai, X.; Yang, Q.; Wang, F.; Wang, F.; Geng, X. The Regulation of ROS- and BECN1-Mediated Autophagy by Human Telomerase Reverse Transcriptase in Glioblastoma. Oxid. Med. Cell. Longev. 2021, 2021, 6636510. [Google Scholar] [CrossRef]
  91. Zhao, X.; Zheng, F.; Li, Y.; Hao, J.; Tang, Z.; Tian, C.; Yang, Q.; Zhu, T.; Diao, C.; Zhang, C.; et al. BPTF Promotes Hepatocellular Carcinoma Growth by Modulating HTERT Signaling and Cancer Stem Cell Traits. Redox Biol. 2019, 20, 427–441. [Google Scholar] [CrossRef]
  92. Park, J.-I.; Venteicher, A.S.; Hong, J.Y.; Choi, J.; Jun, S.; Shkreli, M.; Chang, W.; Meng, Z.; Cheung, P.; Ji, H.; et al. Telomerase Modulates Wnt Signalling by Association with Target Gene Chromatin. Nature 2009, 460, 66–72. [Google Scholar] [CrossRef] [Green Version]
  93. Sanyal, S.; Mondal, P.; Sen, S.; Sengupta, S.; Das, C. SUMO E3 Ligase CBX4 Regulates HTERT-Mediated Transcription of CDH1 and Promotes Breast Cancer Cell Migration and Invasion. Biochem. J. 2020, 477, 3803–3818. [Google Scholar] [CrossRef]
  94. Li, J.; Zhang, N.; Zhang, R.; Sun, L.; Yu, W.; Guo, W.; Gao, Y.; Li, M.; Liu, W.; Liang, P.; et al. CDC5L Promotes HTERT Expression and Colorectal Tumor Growth. Cell. Physiol. Biochem. 2017, 41, 2475–2488. [Google Scholar] [CrossRef] [PubMed]
  95. Yasukawa, M.; Ando, Y.; Yamashita, T.; Matsuda, Y.; Shoji, S.; Morioka, M.S.; Kawaji, H.; Shiozawa, K.; Machitani, M.; Abe, T.; et al. CDK1 Dependent Phosphorylation of HTERT Contributes to Cancer Progression. Nat. Commun. 2020, 11, 1557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Chen, B.-J.; Zeng, S.; Xie, R.; Hu, C.-J.; Wang, S.-M.; Wu, Y.-Y.; Xiao, Y.-F.; Yang, S.-M. HTERT Promotes Gastric Intestinal Metaplasia by Upregulating CDX2 via NF-ΚB Signaling Pathway. Oncotarget 2017, 8, 26969–26978. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Wu, K.-J.; Grandori, C.; Amacker, M.; Simon-Vermot, N.; Polack, A.; Lingner, J.; Dalla-Favera, R. Direct Activation of TERT Transcription by C-MYC. Nat. Genet. 1999, 21, 220–224. [Google Scholar] [CrossRef]
  98. Eldholm, V.; Haugen, A.; Zienolddiny, S. CTCF Mediates the TERT Enhancer-Promoter Interactions in Lung Cancer Cells: Identification of a Novel Enhancer Region Involved in the Regulation of TERT Gene. Int. J. Cancer 2014, 134, 2305–2313. [Google Scholar] [CrossRef]
  99. Zhou, Z.; Li, Y.; Xu, H.; Xie, X.; He, Z.; Lin, S.; Li, R.; Jin, S.; Cui, J.; Hu, H.; et al. An Inducible CRISPR/Cas9 Screen Identifies DTX2 as a Transcriptional Regulator of Human Telomerase. iScience 2022, 25, 103813. [Google Scholar] [CrossRef]
  100. Crowe, D.L.; Nguyen, D.C. Rb and E2F-1 Regulate Telomerase Activity in Human Cancer Cells. Biochim. Biophys. Acta-Gene Struct. Expr. 2001, 1518, 1–6. [Google Scholar] [CrossRef]
  101. Zhang, Y.; Zhang, A.; Shen, C.; Zhang, B.; Rao, Z.; Wang, R.; Yang, S.; Ning, S.; Mao, G.; Fang, D. E2F1 Acts as a Negative Feedback Regulator of C-Myc-Induced HTERT Transcription during Tumorigenesis. Oncol. Rep. 2014, 32, 1273–1280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Lin, C.; Qin, Y.; Zhang, H.; Gao, M.; Wang, Y. EGF Upregulates RFPL3 and HTERT via the MEK Signaling Pathway in Non-small Cell Lung Cancer Cells. Oncol. Rep. 2018, 40, 29–38. [Google Scholar] [CrossRef] [PubMed]
  103. Zhdanov, D.D.; Vasina, D.A.; Grachev, V.A.; Orlova, E.V.; Orlova, V.S.; Pokrovskaya, M.V.; Alexandrova, S.S.; Sokolov, N.N. Alternative Splicing of Telomerase Catalytic Subunit HTERT Generated by Apoptotic Endonuclease EndoG Induces Human CD4+ T Cell Death. Eur. J. Cell Biol. 2017, 96, 653–664. [Google Scholar] [CrossRef] [PubMed]
  104. Grasselli, A.; Nanni, S.; Colussi, C.; Aiello, A.; Benvenuti, V.; Ragone, G.; Moretti, F.; Sacchi, A.; Bacchetti, S.; Gaetano, C.; et al. Estrogen Receptor-α and Endothelial Nitric Oxide Synthase Nuclear Complex Regulates Transcription of Human Telomerase. Circ. Res. 2008, 103, 34–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Akutagawa, O.; Nishi, H.; Kyo, S.; Higuma, C.; Inoue, M.; Isaka, K. Early Growth Response-1 Mediates Up-Regulation of Telomerase in Placenta. Placenta 2007, 28, 920–927. [Google Scholar] [CrossRef]
  106. Yuseran, H.; Hartoyo, E.; Nurseta, T.; Kalim, H. Molecular Docking of Genistein on Estrogen Receptors, Promoter Region of BCLX, Caspase-3, Ki-67, Cyclin D1, and Telomere Activity. J. Taibah Univ. Med. Sci. 2019, 14, 79–87. [Google Scholar] [CrossRef] [PubMed]
  107. Guo, X.; Chen, T.; Chen, S.; Song, C.; Shan, D.; Xu, S.; Xu, S. Case Report: Identification of Multiple TERT and FGFR2 Gene Fusions in a Pineal Region Glioblastoma Case. Front. Oncol. 2021, 11, 739309. [Google Scholar] [CrossRef]
  108. Hu, C.; Ni, Z.; Li, B.; Yong, X.; Yang, X.; Zhang, J.; Zhang, D.; Qin, Y.; Jie, M.; Dong, H.; et al. HTERT Promotes the Invasion of Gastric Cancer Cells by Enhancing FOXO3a Ubiquitination and Subsequent ITGB1 Upregulation. Gut 2017, 66, 31–42. [Google Scholar] [CrossRef]
  109. Xing, X.; Mu, N.; Yuan, X.; Wang, N.; Juhlin, C.C.; Strååt, K.; Larsson, C.; Neo, S.Y.; Xu, D. Downregulation and Hypermethylation of GABPB1 Is Associated with Aggressive Thyroid Cancer Features. Cancers 2022, 14, 1385. [Google Scholar] [CrossRef]
  110. Wang, D.-X.; Zhu, X.-D.; Ma, X.-R.; Wang, L.-B.; Dong, Z.-J.; Lin, R.-R.; Cao, Y.-N.; Zhao, J.-W. Loss of Growth Differentiation Factor 11 Shortens Telomere Length by Downregulating Telomerase Activity. Front. Physiol. 2021, 12, 726345. [Google Scholar] [CrossRef]
  111. Wu, L.; Wang, S.; Tang, B.; Tang, L.; Lei, Y.; Liu, Y.; Yang, M.; Yang, G.; Zhang, D.; Liu, E. Human Telomerase Reverse Transcriptase (HTERT) Synergistic with Sp1 Upregulate Gli1 Expression and Increase Gastric Cancer Invasion and Metastasis. J. Mol. Histol. 2021, 52, 1165–1175. [Google Scholar] [CrossRef]
  112. Wang, K.; Jiang, S.; Huang, A.; Gao, Y.; Peng, B.; Li, Z.; Ma, W.; Songyang, Z.; Zhang, S.; He, M.; et al. GOLPH3 Promotes Cancer Growth by Interacting with STIP1 and Regulating Telomerase Activity in Pancreatic Ductal Adenocarcinoma. Front. Oncol. 2020, 10, 575358. [Google Scholar] [CrossRef]
  113. Lu, H.; Lyu, Y.; Tran, L.; Lan, J.; Xie, Y.; Yang, Y.; Murugan, N.L.; Wang, Y.J.; Semenza, G.L. HIF-1 Recruits NANOG as a Coactivator for TERT Gene Transcription in Hypoxic Breast Cancer Stem Cells. Cell Rep. 2021, 36, 109757. [Google Scholar] [CrossRef] [PubMed]
  114. Choi, S.H.; Cho, K.J.; Yun, S.H.; Jin, B.; Lee, H.Y.; Ro, S.W.; Kim, D.Y.; Ahn, S.H.; Han, K.; Park, J.Y. HKR3 Regulates Cell Cycle through the Inhibition of HTERT in Hepatocellular Carcinoma Cell Lines. J. Cancer 2020, 11, 2442–2452. [Google Scholar] [CrossRef] [PubMed]
  115. Yan, T.; Ooi, W.F.; Qamra, A.; Cheung, A.; Ma, D.; Sundaram, G.M.; Xu, C.; Xing, M.; Poon, L.; Wang, J.; et al. HoxC5 and MiR-615-3p Target Newly Evolved Genomic Regions to Repress HTERT and Inhibit Tumorigenesis. Nat. Commun. 2018, 9, 100. [Google Scholar] [CrossRef] [Green Version]
  116. Sharma, G.G.; Hwang, K.; Pandita, R.K.; Gupta, A.; Dhar, S.; Parenteau, J.; Agarwal, M.; Worman, H.J.; Wellinger, R.J.; Pandita, T.K. Human Heterochromatin Protein 1 Isoforms HP1 Hsα and HP1 Hsβ Interfere with HTERT-Telomere Interactions and Correlate with Changes in Cell Growth and Response to Ionizing Radiation. Mol. Cell. Biol. 2003, 23, 8363–8376. [Google Scholar] [CrossRef] [Green Version]
  117. Chung, S.S.; Wu, Y.; Okobi, Q.; Adekoya, D.; Atefi, M.; Clarke, O.; Dutta, P.; Vadgama, J.V. Proinflammatory Cytokines IL-6 and TNF-α Increased Telomerase Activity through NF-κ B/STAT1/STAT3 Activation, and Withaferin A Inhibited the Signaling in Colorectal Cancer Cells. Med. Inflamm. 2017, 2017, 5958429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Hara, T.; Mizuguchi, M.; Fujii, M.; Nakamura, M. Krüppel-like Factor 2 Represses Transcription of the Telomerase Catalytic Subunit Human Telomerase Reverse Transcriptase (HTERT) in Human T Cells. J. Biol. Chem. 2015, 290, 8758–8763. [Google Scholar] [CrossRef] [Green Version]
  119. Wong, C.-W.; Hou, P.-S.; Tseng, S.-F.; Chien, C.-L.; Wu, K.-J.; Chen, H.-F.; Ho, H.-N.; Kyo, S.; Teng, S.-C. Krüppel-Like Transcription Factor 4 Contributes to Maintenance of Telomerase Activity in Stem Cells. Stem Cells 2010, 28, 1510–1517. [Google Scholar] [CrossRef]
  120. Zhang, C.; Song, C.; Liu, T.; Tang, R.; Chen, M.; Gao, F.; Xiao, B.; Qin, G.; Shi, F.; Li, W.; et al. KMT2A Promotes Melanoma Cell Growth by Targeting HTERT Signaling Pathway. Cell Death Dis. 2017, 8, e2940. [Google Scholar] [CrossRef] [Green Version]
  121. Briatore, F.; Barrera, G.; Pizzimenti, S.; Toaldo, C.; Della Casa, C.; Laurora, S.; Pettazzoni, P.; Dianzani, M.U.; Ferrero, D. Increase of Telomerase Activity and HTERT Expression in Myelodysplastic Syndromes. Cancer Biol. Ther. 2009, 8, 883–889. [Google Scholar] [CrossRef] [Green Version]
  122. Liu, H.; Liu, Q.; Ge, Y.; Zhao, Q.; Zheng, X.; Zhao, Y. HTERT Promotes Cell Adhesion and Migration Independent of Telomerase Activity. Sci. Rep. 2016, 6, 22886. [Google Scholar] [CrossRef] [Green Version]
  123. Tang, Y.-L.; Sun, X.; Huang, L.-B.; Liu, X.-J.; Qin, G.; Wang, L.-N.; Zhang, X.-L.; Ke, Z.-Y.; Luo, J.-S.; Liang, C.; et al. Melatonin Inhibits MLL-Rearranged Leukemia via RBFOX3/HTERT and NF-ΚB/COX-2 Signaling Pathways. Cancer Lett. 2019, 443, 167–178. [Google Scholar] [CrossRef]
  124. Karlsen, T.R.; Olsen, M.B.; Kong, X.Y.; Yang, K.; Quiles-Jiménez, A.; Kroustallaki, P.; Holm, S.; Lines, G.T.; Aukrust, P.; Skarpengland, T.; et al. NEIL3-Deficient Bone Marrow Displays Decreased Hematopoietic Capacity and Reduced Telomere Length. Biochem. Biophys. Rep. 2022, 29, 101211. [Google Scholar] [CrossRef] [PubMed]
  125. Aravindan, N.; Veeraraghavan, J.; Madhusoodhanan, R.; Herman, T.S.; Natarajan, M. Curcumin Regulates Low-Linear Energy Transfer γ-Radiation-Induced NFκB-Dependent Telomerase Activity in Human Neuroblastoma Cells. Int. J. Radiat. Oncol. 2011, 79, 1206–1215. [Google Scholar] [CrossRef] [Green Version]
  126. Gizard, F.; Heywood, E.B.; Findeisen, H.M.; Zhao, Y.; Jones, K.L.; Cudejko, C.; Post, G.R.; Staels, B.; Bruemmer, D. Telomerase Activation in Atherosclerosis and Induction of Telomerase Reverse Transcriptase Expression by Inflammatory Stimuli in Macrophages. Arterioscler. Thromb. Vasc. Biol. 2011, 31, 245–252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Gewin, L. Identification of a Novel Telomerase Repressor That Interacts with the Human Papillomavirus Type-16 E6/E6-AP Complex. Genes Dev. 2004, 18, 2269–2282. [Google Scholar] [CrossRef] [Green Version]
  128. Saha, D.; Singh, A.; Hussain, T.; Srivastava, V.; Sengupta, S.; Kar, A.; Dhapola, P.; Dhople, V.; Ummanni, R.; Chowdhury, S. Epigenetic Suppression of Human Telomerase (HTERT) Is Mediated by the Metastasis Suppressor NME2 in a G-Quadruplex–Dependent Fashion. J. Biol. Chem. 2017, 292, 15205–15215. [Google Scholar] [CrossRef] [Green Version]
  129. Sayed, M.E.; Yuan, L.; Robin, J.D.; Tedone, E.; Batten, K.; Dahlson, N.; Wright, W.E.; Shay, J.W.; Ludlow, A.T. NOVA1 Directs PTBP1 to HTERT Pre-MRNA and Promotes Telomerase Activity in Cancer Cells. Oncogene 2019, 38, 2937–2952. [Google Scholar] [CrossRef] [Green Version]
  130. Zhao, T.; Zhao, C.; Lu, Y.; Lin, J.; Tian, Y.; Ma, Y.; Li, J.; Zhang, H.; Yan, W.; Jiao, P.; et al. Noxa and Puma Genes Regulated by HTERT Promoter Can Mitigate Growth and Induce Apoptosis in Hepatocellular Carcinoma Mouse Model. J. Cancer 2022, 13, 2001–2013. [Google Scholar] [CrossRef] [PubMed]
  131. Dong, H.; Xia, Y.; Jin, S.; Xue, C.; Wang, Y.; Hu, R.; Jiang, H. Nrf2 Attenuates Ferroptosis-Mediated IIR-ALI by Modulating TERT and SLC7A11. Cell Death Dis. 2021, 12, 1027. [Google Scholar] [CrossRef] [PubMed]
  132. Romanova, L.; Kellner, S.; Katoku-Kikyo, N.; Kikyo, N. Novel Role of Nucleostemin in the Maintenance of Nucleolar Architecture and Integrity of Small Nucleolar Ribonucleoproteins and the Telomerase Complex. J. Biol. Chem. 2009, 284, 26685–26694. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Beitzinger, M.; Oswald, C.; Beinoraviciute-Kellner, R.; Stiewe, T. Regulation of Telomerase Activity by the P53 Family Member P73. Oncogene 2006, 25, 813–826. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Bougel, S.; Renaud, S.; Braunschweig, R.; Loukinov, D.; Morse, H.C., III; Bosman, F.T.; Lobanenkov, V.; Benhattar, J. PAX5 Activates the Transcription of the Human Telomerase Reverse Transcriptase Gene in B Cells. J. Pathol. 2010, 220, 87–96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Chen, Y.-J.; Campbell, H.G.; Wiles, A.K.; Eccles, M.R.; Reddel, R.R.; Braithwaite, A.W.; Royds, J.A. PAX8 Regulates Telomerase Reverse Transcriptase and Telomerase RNA Component in Glioma. Cancer Res. 2008, 68, 5724–5732. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Hu, J.; Hwang, S.S.; Liesa, M.; Gan, B.; Sahin, E.; Jaskelioff, M.; Ding, Z.; Ying, H.; Boutin, A.T.; Zhang, H.; et al. Antitelomerase Therapy Provokes ALT and Mitochondrial Adaptive Mechanisms in Cancer. Cell 2012, 148, 651–663. [Google Scholar] [CrossRef] [Green Version]
  137. Ho, S.-T.; Jin, R.; Cheung, D.H.-C.; Huang, J.-J.; Shaw, P.-C. The PinX1/NPM Interaction Associates with HTERT in Early-S Phase and Facilitates Telomerase Activation. Cell Biosci. 2019, 9, 47. [Google Scholar] [CrossRef]
  138. Ohira, T.; Kojima, H.; Kuroda, Y.; Aoki, S.; Inaoka, D.; Osaki, M.; Wanibuchi, H.; Okada, F.; Oshimura, M.; Kugoh, H. PITX1 Protein Interacts with ZCCHC10 to Regulate HTERT MRNA Transcription. PLoS ONE 2019, 14, e0217605. [Google Scholar] [CrossRef] [Green Version]
  139. Zhang, Q.; Feng, W.; Wang, Q.; Wang, J.; Chai, L.; Chen, Y.; Wang, Y.; Liu, J.; Li, M.; Xie, X. PPARγ Activation Inhibits PDGF-Induced Pulmonary Artery Smooth Muscle Cell Proliferation and Migration by Modulating TERT. Biomed. Pharmacother. 2022, 152, 113233. [Google Scholar] [CrossRef]
  140. Luo, C.; Zhu, X.; Luo, Q.; Bu, F.; Huang, C.; Zhu, J.; Zhao, J.; Zhang, W.; Lin, K.; Hu, C.; et al. RBFOX3 Promotes Gastric Cancer Growth and Progression by Activating HTERT Signaling. Front. Oncol. 2020, 10, 1044. [Google Scholar] [CrossRef]
  141. Liu, T.; Li, W.; Lu, W.; Chen, M.; Luo, M.; Zhang, C.; Li, Y.; Qin, G.; Shi, D.; Xiao, B.; et al. RBFOX3 Promotes Tumor Growth and Progression via HTERT Signaling and Predicts a Poor Prognosis in Hepatocellular Carcinoma. Theranostics 2017, 7, 3138–3154. [Google Scholar] [CrossRef]
  142. Zohud, B.A.; Guo, P.; Zohud, B.A.; Li, F.; Hao, J.J.; Shan, X.; Yu, W.; Guo, W.; Qin, Y.; Cai, X. Importin 13 Promotes NSCLC Progression by Mediating RFPL3 Nuclear Translocation and HTERT Expression Upregulation. Cell Death Dis. 2020, 11, 879. [Google Scholar] [CrossRef]
  143. Liu, Y.-B.; Mei, Y.; Long, J.; Zhang, Y.; Hu, D.-L.; Zhou, H.-H. RIF1 Promotes Human Epithelial Ovarian Cancer Growth and Progression via Activating Human Telomerase Reverse Transcriptase Expression. J. Exp. Clin. Cancer Res. 2018, 37, 182. [Google Scholar] [CrossRef] [PubMed]
  144. Zhang, W.; Veisaga, M.L.; Barbieri, M.A. Role of RIN1 on Telomerase Activity Driven by EGF-Ras Mediated Signaling in Breast Cancer. Exp. Cell Res. 2020, 396, 112318. [Google Scholar] [CrossRef] [PubMed]
  145. You, Y.; Sun, X.; Xiao, J.; Chen, Y.; Chen, X.; Pang, J.; Mi, J.; Tang, Y.; Liu, Q.; Ling, W. Inhibition of S-Adenosylhomocysteine Hydrolase Induces Endothelial Senescence via HTERT Downregulation. Atherosclerosis 2022, 353, 1–10. [Google Scholar] [CrossRef] [PubMed]
  146. Parulekar, A.; Choksi, A.; Taye, N.; Totakura, K.V.S.; Firmal, P.; Kundu, G.C.; Chattopadhyay, S. SMAR1 Suppresses the Cancer Stem Cell Population via HTERT Repression in Colorectal Cancer Cells. Int. J. Biochem. Cell Biol. 2021, 141, 106085. [Google Scholar] [CrossRef] [PubMed]
  147. Won, J.; Yim, J.; Kim, T.K. Sp1 and Sp3 Recruit Histone Deacetylase to Repress Transcription of Human Telomerase Reverse Transcriptase (HTERT) Promoter in Normal Human Somatic Cells. J. Biol. Chem. 2002, 277, 38230–38238. [Google Scholar] [CrossRef] [Green Version]
  148. Dratwa, M.; Wysoczanska, B.; Brankiewicz, W.; Stachowicz-Suhs, M.; Wietrzyk, J.; Matkowski, R.; Ekiert, M.; Szelachowska, J.; Maciejczyk, A.; Szajewski, M.; et al. Relationship between Telomere Length, TERT Genetic Variability and TERT, TP53, SP1, MYC Gene Co-Expression in the Clinicopathological Profile of Breast Cancer. Int. J. Mol. Sci. 2022, 23, 5164. [Google Scholar] [CrossRef] [PubMed]
  149. Cheng, D.; Zhao, Y.; Wang, S.; Jia, W.; Kang, J.; Zhu, J. Human Telomerase Reverse Transcriptase (HTERT) Transcription Requires Sp1/Sp3 Binding to the Promoter and a Permissive Chromatin Environment. J. Biol. Chem. 2015, 290, 30193–30203. [Google Scholar] [CrossRef] [Green Version]
  150. Diao, C.; Guo, P.; Yang, W.; Sun, Y.; Liao, Y.; Yan, Y.; Zhao, A.; Cai, X.; Hao, J.; Hu, S.; et al. SPT6 Recruits SND1 to Co-activate Human Telomerase Reverse Transcriptase to Promote Colon Cancer Progression. Mol. Oncol. 2021, 15, 1180–1202. [Google Scholar] [CrossRef] [PubMed]
  151. Chung, S.S.; Aroh, C.; Vadgama, J.V. Constitutive Activation of STAT3 Signaling Regulates HTERT and Promotes Stem Cell-Like Traits in Human Breast Cancer Cells. PLoS ONE 2013, 8, e83971. [Google Scholar] [CrossRef]
  152. Chang, W.-T.; Lin, Y.-C.; Hong, C.-S.; Huang, P.-S.; Lin, Y.-W.; Chen, Z.-C.; Lin, T.-H.; Chao, T.-H. Effects of STAT3 on Aging-Dependent Neovascularization Impairment Following Limb Ischemia: From Bedside to Bench. Aging 2022, 14, 4897–4913. [Google Scholar] [CrossRef]
  153. Yamada, O.; Ozaki, K.; Akiyama, M.; Kawauchi, K. JAK–STAT and JAK–PI3K–MTORC1 Pathways Regulate Telomerase Transcriptionally and Posttranslationally in ATL Cells. Mol. Cancer Ther. 2012, 11, 1112–1121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Endoh, T.; Tsuji, N.; Asanuma, K.; Yagihashi, A.; Watanabe, N. Survivin Enhances Telomerase Activity via Up-Regulation of Specificity Protein 1- and c-Myc-Mediated Human Telomerase Reverse Transcriptase Gene Transcription. Exp. Cell Res. 2005, 305, 300–311. [Google Scholar] [CrossRef]
  155. Miao, B.; Zhang, C.; Stroh, N.; Brenner, L.; Hufnagel, K.; Hoheisel, J.D.; Bandapalli, O.R. Transcription Factor TFE3 Enhances Cell Cycle and Cancer Progression by Binding to the HTERT Promoter. Cancer Commun. 2021, 41, 1423–1426. [Google Scholar] [CrossRef]
  156. Burgess, J.K.; Ketheson, A.; Faiz, A.; Limbert Rempel, K.A.; Oliver, B.G.; Ward, J.P.T.; Halayko, A.J. Phenotype and Functional Features of Human Telomerase Reverse Transcriptase Immortalized Human Airway Smooth Muscle Cells from Asthmatic and Non-Asthmatic Donors. Sci. Rep. 2018, 8, 805. [Google Scholar] [CrossRef] [Green Version]
  157. Li, Y.; Zhang, N.; Ma, C.; Xu, W.; Jin, G.; Zheng, Y.; Zhang, L.; Liu, B.; Gao, C.; Liu, S. The Overexpression of Tipe2 in CRC Cells Suppresses Survival While Endogenous Tipe2 Accelerates AOM/DSS Induced-Tumor Initiation. Cell Death Dis. 2021, 12, 1001. [Google Scholar] [CrossRef]
  158. Deng, T.; Huang, Y.; Weng, K.; Lin, S.; Li, Y.; Shi, G.; Chen, Y.; Huang, J.; Liu, D.; Ma, W.; et al. TOE1 Acts as a 3′ Exonuclease for Telomerase RNA and Regulates Telomere Maintenance. Nucleic Acids Res. 2019, 47, 391–405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Olbertova, H.; Plevova, K.; Pavlova, S.; Malcikova, J.; Kotaskova, J.; Stranska, K.; Spunarova, M.; Trbusek, M.; Navrkalova, V.; Dvorackova, B.; et al. Evolution of TP53 Abnormalities during CLL Disease Course Is Associated with Telomere Length Changes. BMC Cancer 2022, 22, 137. [Google Scholar] [CrossRef] [PubMed]
  160. Agarwal, N.; Rinaldetti, S.; Cheikh, B.B.; Zhou, Q.; Hass, E.P.; Jones, R.T.; Joshi, M.; LaBarbera, D.V.; Knott, S.R.V.; Cech, T.R.; et al. TRIM28 Is a Transcriptional Activator of the Mutant TERT Promoter in Human Bladder Cancer. Proc. Natl. Acad. Sci. USA 2021, 118, e2102423118. [Google Scholar] [CrossRef]
  161. Che, Y.; Li, Y.; Zheng, F.; Zou, K.; Li, Z.; Chen, M.; Hu, S.; Tian, C.; Yu, W.; Guo, W.; et al. TRIP4 Promotes Tumor Growth and Metastasis and Regulates Radiosensitivity of Cervical Cancer by Activating MAPK, PI3K/AKT, and HTERT Signaling. Cancer Lett. 2019, 452, 1–13. [Google Scholar] [CrossRef] [PubMed]
  162. Goueli, B.S.; Janknecht, R. Regulation of Telomerase Reverse Transcriptase Gene Activity by Upstream Stimulatory Factor. Oncogene 2003, 22, 8042–8047. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Yago, M.; Ohki, R.; Hatakeyama, S.; Fujita, T.; Ishikawa, F. Variant Forms of Upstream Stimulatory Factors (USFs) Control the Promoter Activity of HTERT, the Human Gene Encoding the Catalytic Subunit of Telomerase. FEBS Lett. 2002, 520, 40–46. [Google Scholar] [CrossRef] [Green Version]
  164. Chang, J.T.-C.; Yang, H.-T.; Wang, T.-C.V.; Cheng, A.-J. Upstream Stimulatory Factor (USF) as a Transcriptional Suppressor of Human Telomerase Reverse Transcriptase (HTERT) in Oral Cancer Cells. Mol. Carcinog. 2005, 44, 183–192. [Google Scholar] [CrossRef] [PubMed]
  165. Yu, P.; Shen, X.; Yang, W.; Zhang, Y.; Liu, C.; Huang, T. ZEB1 Stimulates Breast Cancer Growth by Up-Regulating HTERT Expression. Biochem. Biophys. Res. Commun. 2018, 495, 2505–2511. [Google Scholar] [CrossRef] [PubMed]
  166. Asfour, I.A.; Fayek, M.H.; El-kourashy, S.A.-E.A.; Youssef, S.R.; El-Gohary, G.M.T.; Mohamed, O.F. Correlation of Telomerase Activity to Apoptosis and Survival in Adult Acute Lymphoblastic Leukemia: An Egyptian Single-Center Study. Ann. Hematol. 2008, 87, 213–221. [Google Scholar] [CrossRef]
  167. Rafat, A.; Dizaji Asl, K.; Mazloumi, Z.; Movassaghpour, A.A.; Farahzadi, R.; Nejati, B.; Nozad Charoudeh, H. Telomerase-based Therapies in Haematological Malignancies. Cell Biochem. Funct. 2022, 40, 127–140. [Google Scholar] [CrossRef]
  168. Porika, M.; Tippani, R.; Saretzki, G.C. CRISPR/Cas: A New Tool in the Research of Telomeres and Telomerase as Well as a Novel Form of Cancer Therapy. Int. J. Mol. Sci. 2022, 23, 3002. [Google Scholar] [CrossRef]
  169. Yik, M.Y.; Azlan, A.; Rajasegaran, Y.; Rosli, A.; Yusoff, N.M.; Moses, E.J. Mechanism of Human Telomerase Reverse Transcriptase (HTERT) Regulation and Clinical Impacts in Leukemia. Genes 2021, 12, 1188. [Google Scholar] [CrossRef]
  170. Weng, N.-P.; Granger, L.; Hodes, R.J. Telomere Lengthening and Telomerase Activation during Human B Cell Differentiation. Proc. Natl. Acad. Sci. USA 1997, 94, 10827–10832. [Google Scholar] [CrossRef] [Green Version]
  171. Son, N.H.; Joyce, B.; Hieatt, A.; Chrest, F.J.; Yanovski, J.; Weng, N. Stable Telomere Length and Telomerase Expression from Naive to Memory B-Lymphocyte Differentiation. Mech. Ageing Dev. 2003, 124, 427–432. [Google Scholar] [CrossRef]
  172. Igarashi, H.; Sakaguchi, N. Telomerase Activity Is Induced in Human Peripheral B Lymphocytes by the Stimulation to Antigen Receptor. Blood 1997, 89, 1299–1307. [Google Scholar] [CrossRef]
  173. Hu, B.T.; Insel, R.A. Up-Regulation of Telomerase in Human B Lymphocytes Occurs Independently of Cellular Proliferation and with Expression of the Telomerase Catalytic Subunit. Eur. J. Immunol. 1999, 29, 3745–3753. [Google Scholar] [CrossRef]
  174. Weng, N. Regulation of Telomerase Expression in Human Lymphocytes. Springer Semin. Immunopathol. 2002, 24, 23–33. [Google Scholar] [CrossRef]
  175. Bienz, M.; Ramdani, S.; Knecht, H. Molecular Pathogenesis of Hodgkin Lymphoma: Past, Present, Future. Int. J. Mol. Sci. 2020, 21, 6623. [Google Scholar] [CrossRef]
  176. Ackermann, S.; Fischer, M. Telomere Maintenance in Pediatric Cancer. Int. J. Mol. Sci. 2019, 20, 5836. [Google Scholar] [CrossRef] [Green Version]
  177. Capraro, V.; Zane, L.; Poncet, D.; Perol, D.; Galia, P.; Preudhomme, C.; Bonnefoy-Berard, N.; Gilson, E.; Thomas, X.; El-Hamri, M. Telomere Deregulations Possess Cytogenetic, Phenotype, and Prognostic Specificities in Acute Leukemias. Exp. Hematol. 2011, 39, 195–202.e2. [Google Scholar] [CrossRef] [PubMed]
  178. Wang, Y.; Fang, M.; Sun, X.; Sun, J. Telomerase Activity and Telomere Length in Acute Leukemia: Correlations with Disease Progression, Subtypes and Overall Survival. Int. J. Lab. Hematol. 2010, 32, 230–238. [Google Scholar] [CrossRef]
  179. Eskandari, E.; Hashemi, M.; Naderi, M.; Bahari, G.; Safdari, V.; Taheri, M. Leukocyte Telomere Length Shortening, HTERT Genetic Polymorphisms and Risk of Childhood Acute Lymphoblastic Leukemia. Asian Pac. J. Cancer Prev. 2018, 19, 1515–1521. [Google Scholar] [CrossRef] [PubMed]
  180. Chien, W.W.; Catallo, R.; Chebel, A.; Baranger, L.; Thomas, X.; Béné, M.-C.; Gerland, L.M.; Schmidt, A.; Beldjord, K.; Klein, N.; et al. The P16INK4A/PRb Pathway and Telomerase Activity Define a Subgroup of Ph+ Adult Acute Lymphoblastic Leukemia Associated with Inferior Outcome. Leuk. Res. 2015, 39, 453–461. [Google Scholar] [CrossRef]
  181. Chai, J.H.; Zhang, Y.; Tan, W.H.; Chng, W.J.; Li, B.; Wang, X. Regulation of HTERT by BCR-ABL at Multiple Levels in K562 Cells. BMC Cancer 2011, 11, 512. [Google Scholar] [CrossRef] [Green Version]
  182. Cogulu, O.; Kosova, B.; Gunduz, C.; Karaca, E.; Aksoylar, S.; Erbay, A.; Karapinar, D.; Vergin, C.; Vural, F.; Tombuloglu, M.; et al. The Evaluation of HTERT MRNA Expression in Acute Leukemia Children and 2 Years Follow-up of 40 Cases. Int. J. Hematol. 2008, 87, 276–283. [Google Scholar] [CrossRef]
  183. Borssén, M.; Cullman, I.; Norén-Nyström, U.; Sundström, C.; Porwit, A.; Forestier, E.; Roos, G. HTERT Promoter Methylation and Telomere Length in Childhood Acute Lymphoblastic Leukemia—Associations with Immunophenotype and Cytogenetic Subgroup. Exp. Hematol. 2011, 39, 1144–1151. [Google Scholar] [CrossRef] [PubMed]
  184. Zobeck, M.; Bernhardt, M.B.; Kamdar, K.Y.; Rabin, K.R.; Lupo, P.J.; Scheurer, M.E. Novel and Replicated Clinical and Genetic Risk Factors for Toxicity from High-dose Methotrexate in Pediatric Acute Lymphoblastic Leukemia. Pharmacother. J. Hum. Pharmacol. Drug Ther. 2023, 00, 1–10. [Google Scholar] [CrossRef] [PubMed]
  185. Popov, A.; Tsaur, G.; Permikin, Z.; Henze, G.; Verzhbitskaya, T.; Plekhanova, O.; Nokhrina, E.; Valochnik, A.; Sibiryakov, P.; Zerkalenkova, E.; et al. Genetic Characteristics and Treatment Outcome in Infants with KMT2A Germline B-cell Precursor Acute Lymphoblastic Leukemia: Results of MLL-Baby Protocol. Pediatr. Blood Cancer 2023, 70, e30204. [Google Scholar] [CrossRef]
  186. Safavi, S.; Olsson, L.; Biloglav, A.; Veerla, S.; Blendberg, M.; Tayebwa, J.; Behrendtz, M.; Castor, A.; Hansson, M.; Johansson, B.; et al. Genetic and Epigenetic Characterization of Hypodiploid Acute Lymphoblastic Leukemia. Oncotarget 2015, 6, 42793–42802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Hu, Q.; Chen, X.; Liu, S.; Wen, R.; Yuan, X.; Xu, D.; Liu, G.; Wen, F. Methylation of CDKN2B CpG Islands Is Associated with Upregulated Telomerase Activity in Children with Acute Lymphoblastic Leukemia. Oncol. Lett. 2017, 13, 2115–2120. [Google Scholar] [CrossRef] [Green Version]
  188. Assi, R.; Mahfouz, R.; Owen, R.; Gunthorpe, M.; Chehab, F.F.; Bazarbachi, A. PAX5, NOTCH3, CBFB, and ACD Drive an Activated RAS Pathway and Monosomy 7 to B-ALL and AML in Donor Cell Leukemia. Bone Marrow Transplant. 2019, 54, 1124–1128. [Google Scholar] [CrossRef] [PubMed]
  189. Spinella, J.-F.; Cassart, P.; Garnier, N.; Rousseau, P.; Drullion, C.; Richer, C.; Ouimet, M.; Saillour, V.; Healy, J.; Autexier, C.; et al. A Novel Somatic Mutation in ACD Induces Telomere Lengthening and Apoptosis Resistance in Leukemia Cells. BMC Cancer 2015, 15, 621. [Google Scholar] [CrossRef] [Green Version]
  190. Zia, S.; Khan, N.; Tehreem, K.; Rehman, N.; Sami, R.; Baty, R.S.; Tayeb, F.J.; Almashjary, M.N.; Alsubhi, N.H.; Alrefaei, G.I.; et al. Transcriptomic Analysis of Conserved Telomere Maintenance Component 1 (CTC1) and Its Association with Leukemia. J. Clin. Med. 2022, 11, 5780. [Google Scholar] [CrossRef] [PubMed]
  191. Sheng, X.; Zhang, L.; Luo, D.; Tong, N.; Wang, M.; Fang, Y.; Li, J.; Zhang, Z. A Common Variant near TERC and Telomere Length Are Associated with Susceptibility to Childhood Acute Lymphoblastic Leukemia in Chinese. Leuk. Lymphoma 2012, 53, 1688–1692. [Google Scholar] [CrossRef]
  192. Reddy, A.; Espinoza, I.; Cole, D.; Schallheim, J.; Poosarla, T.; Bhanat, E.; Zhou, Y.; Zabaleta, J.; Megason, G.; Gomez, C.R. Genetic Mutations in B-Acute Lymphoblastic Leukemia Among African American and European American Children. Clin. Lymphoma Myeloma Leuk. 2018, 18, e501–e508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Zvereva, M.I.; Zatsepin, T.S.; Azhibek, D.M.; Shubernetskaya, O.S.; Shpanchenko, O.V.; Dontsova, O.A. Oligonucleotide Inhibitors of Telomerase: Prospects for Anticancer Therapy and Diagnostics. Biochemistry 2015, 80, 251–259. [Google Scholar] [CrossRef]
  194. Zhou, C.; Liu, S. Evaluation of the Efficacy of MST-312, as a Telomerase Inhibitor, in the Treatment of Patients with Multiple Myeloma after Stem Cell Transplantation. Cell. Mol. Biol. 2022, 67, 115–120. [Google Scholar] [CrossRef] [PubMed]
  195. Andrade da Mota, T.H.; Reis Guimarães, A.F.; Silva de Carvalho, A.É.; Saldanha-de Araujo, F.; Pinto de Faria Lopes, G.; Pittella-Silva, F.; do Amaral Rabello, D.; Madureira de Oliveira, D. Effects of in Vitro Short- and Long-Term Treatment with Telomerase Inhibitor in U-251 Glioma Cells. Tumor Biol. 2021, 43, 327–340. [Google Scholar] [CrossRef]
  196. Gurung, R.L.; Lim, S.N.; Low, G.K.M.; Hande, M.P. MST-312 Alters Telomere Dynamics, Gene Expression Profiles and Growth in Human Breast Cancer Cells. Lifestyle Genom. 2014, 7, 283–298. [Google Scholar] [CrossRef]
  197. Sajed, H.; Sahebkar, A.; Iranshahi, M. Zataria multiflora Boiss. (Shirazi Thyme)—An Ancient Condiment with Modern Pharmaceutical Uses. J. Ethnopharmacol. 2013, 145, 686–698. [Google Scholar] [CrossRef]
  198. Lashkari, M.; Fatemi, A.; Valandani, H.M.; Khalilabadi, R.M. Promising Anti-Leukemic Effect of Zataria multiflora Extract in Combination with Doxorubicin to Combat Acute Lymphoblastic Leukemia Cells (Nalm-6) (In Vitro and In Silico). Sci. Rep. 2022, 12, 12657. [Google Scholar] [CrossRef]
  199. Habibi, E.; Shokrzadeh, M.; Ahmadi, A.; Chabra, A.; Naghshvar, F.; Haghi-Aminjan, H.; Salehi, F. Pulmonoprotective Action of Zataria multiflora Ethanolic Extract on Cyclophosphamide-Induced Oxidative Lung Toxicity in Mice. Chin. J. Integr. Med. 2020, 26, 754–761. [Google Scholar] [CrossRef]
  200. Altamura, G.; degli Uberti, B.; Galiero, G.; De Luca, G.; Power, K.; Licenziato, L.; Maiolino, P.; Borzacchiello, G. The Small Molecule BIBR1532 Exerts Potential Anti-Cancer Activities in Preclinical Models of Feline Oral Squamous Cell Carcinoma Through Inhibition of Telomerase Activity and Down-Regulation of TERT. Front. Vet. Sci. 2021, 7, 620776. [Google Scholar] [CrossRef] [PubMed]
  201. El-Daly, H.; Kull, M.; Zimmermann, S.; Pantic, M.; Waller, C.F.; Martens, U.M. Selective Cytotoxicity and Telomere Damage in Leukemia Cells Using the Telomerase Inhibitor BIBR1532. Blood 2005, 105, 1742–1749. [Google Scholar] [CrossRef]
  202. Pourbagheri-Sigaroodi, A.; Bashash, D.; Safaroghli-Azar, A.; Farshi-Paraasghari, M.; Momeny, M.; Mansoor, F.N.; Ghaffari, S.H. Contributory Role of MicroRNAs in Anti-Cancer Effects of Small Molecule Inhibitor of Telomerase (BIBR1532) on Acute Promyelocytic Leukemia Cell Line. Eur. J. Pharmacol. 2019, 846, 49–62. [Google Scholar] [CrossRef]
  203. Bashash, D.; Ghaffari, S.H.; Zaker, F.; Hezave, K.; Kazerani, M.; Ghavamzadeh, A.; Alimoghaddam, K.; Mosavi, S.A.; Gharehbaghian, A.; Vossough, P. Direct Short-Term Cytotoxic Effects of BIBR 1532 on Acute Promyelocytic Leukemia Cells Through Induction of P21 Coupled with Downregulation of c-Myc and HTERT Transcription. Cancer Invest. 2012, 30, 57–64. [Google Scholar] [CrossRef] [PubMed]
  204. Barwe, S.P.; Huang, F.; Kolb, E.A.; Gopalakrishnapillai, A. Imetelstat Induces Leukemia Stem Cell Death in Pediatric Acute Myeloid Leukemia Patient-Derived Xenografts. J. Clin. Med. 2022, 11, 1923. [Google Scholar] [CrossRef] [PubMed]
  205. Marian, C.O.; Cho, S.K.; Mcellin, B.M.; Maher, E.A.; Hatanpaa, K.J.; Madden, C.J.; Mickey, B.E.; Wright, W.E.; Shay, J.W.; Bachoo, R.M. The Telomerase Antagonist, Imetelstat, Efficiently Targets Glioblastoma Tumor-Initiating Cells Leading to Decreased Proliferation and Tumor Growth. Clin. Cancer Res. 2010, 16, 154–163. [Google Scholar] [CrossRef] [Green Version]
  206. Pilco-Ferreto, N.; Calaf, G.M. Influence of Doxorubicin on Apoptosis and Oxidative Stress in Breast Cancer Cell Lines. Int. J. Oncol. 2016, 49, 753–762. [Google Scholar] [CrossRef] [Green Version]
  207. Rawat, P.S.; Jaiswal, A.; Khurana, A.; Bhatti, J.S.; Navik, U. Doxorubicin-Induced Cardiotoxicity: An Update on the Molecular Mechanism and Novel Therapeutic Strategies for Effective Management. Biomed. Pharmacother. 2021, 139, 111708. [Google Scholar] [CrossRef] [PubMed]
  208. Ghasemimehr, N.; Farsinejad, A.; Mirzaee Khalilabadi, R.; Yazdani, Z.; Fatemi, A. The Telomerase Inhibitor MST-312 Synergistically Enhances the Apoptotic Effect of Doxorubicin in Pre-B Acute Lymphoblastic Leukemia Cells. Biomed. Pharmacother. 2018, 106, 1742–1750. [Google Scholar] [CrossRef] [PubMed]
  209. Wu, L.; Fidan, K.; Um, J.-Y.; Ahn, K.S. Telomerase: Key Regulator of Inflammation and Cancer. Pharmacol. Res. 2020, 155, 104726. [Google Scholar] [CrossRef] [PubMed]
  210. Pascolo, E.; Wenz, C.; Lingner, J.; Hauel, N.; Priepke, H.; Kauffmann, I.; Garin-Chesa, P.; Rettig, W.J.; Damm, K.; Schnapp, A. Mechanism of Human Telomerase Inhibition by BIBR1532, a Synthetic, Non-Nucleosidic Drug Candidate. J. Biol. Chem. 2002, 277, 15566–15572. [Google Scholar] [CrossRef] [Green Version]
  211. Bashash, D.; Zareii, M.; Safaroghli-Azar, A.; Omrani, M.D.; Ghaffari, S.H. Inhibition of Telomerase Using BIBR1532 Enhances Doxorubicin-Induced Apoptosis in Pre-B Acute Lymphoblastic Leukemia Cells. Hematology 2017, 22, 330–340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Mengual Gomez, D.L.; Armando, R.G.; Cerrudo, C.S.; Ghiringhelli, P.D.; Gomez, D.E. Telomerase as a Cancer Target. Development of New Molecules. Curr. Top. Med. Chem. 2016, 16, 2432–2440. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Relitti, N.; Saraswati, A.P.; Federico, S.; Khan, T.; Brindisi, M.; Zisterer, D.; Brogi, S.; Gemma, S.; Butini, S.; Campiani, G. Telomerase-Based Cancer Therapeutics: A Review on Their Clinical Trials. Curr. Top. Med. Chem. 2020, 20, 433–457. [Google Scholar] [CrossRef] [PubMed]
  214. Mizukoshi, E.; Kaneko, S. Telomerase-Targeted Cancer Immunotherapy. Int. J. Mol. Sci. 2019, 20, 1823. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Telomerase representation. Dyskerin complex (NHP2, NOP10 and DKC) binds to hTR through its ACA domain. TCAB1 binds to TERT and to hTR. The template region of hTR binds to the telomeric 3′ -end strand. Shelterin binds to telomeric repeat region. TRF2 interacts with RAP1 and TRF1, binding directly to the telomeric DNA and to TIN2, which also binds to TPP1. TPP1 interacts with POT1, which is responsible for recruiting telomerase to telomeres through the TEN domain of TERT.
Figure 1. Telomerase representation. Dyskerin complex (NHP2, NOP10 and DKC) binds to hTR through its ACA domain. TCAB1 binds to TERT and to hTR. The template region of hTR binds to the telomeric 3′ -end strand. Shelterin binds to telomeric repeat region. TRF2 interacts with RAP1 and TRF1, binding directly to the telomeric DNA and to TIN2, which also binds to TPP1. TPP1 interacts with POT1, which is responsible for recruiting telomerase to telomeres through the TEN domain of TERT.
Genes 14 00691 g001
Figure 2. Model of telomere regulation in B cells. The green color represents telomere length and the blue color the chromosomes. The germinal center has greater telomere length than naive and memory cells.
Figure 2. Model of telomere regulation in B cells. The green color represents telomere length and the blue color the chromosomes. The germinal center has greater telomere length than naive and memory cells.
Genes 14 00691 g002
Table 1. Telomerase inhibitors.
Table 1. Telomerase inhibitors.
CompoundsChemical StructuresType of TherapyMain FindingsReferences
N,N′-1,3-Phenylenebis-[2,3-dihydroxy-benzamide]
(MST-312)
Genes 14 00691 i001Combined with doxorubicinMST-312 inhibits the progress of multiple myeloma by inhibiting the telomerase activity of this cells.
Monotherapy long-term exposure to the MST-312 in U251 cells resulted in the induction of cell adaptations with possible negative clinical implications.
MST-312 alters telomere dynamics, gene expression profiles and growth in human breast cancer cells
[194,195,196]
Zataria multiflora extract (ZME)Chemical structures of main volatile and non-volatile constituents are in Sajed, Sahebkar and Iranshahi works [197]Combined with doxorubicinPulmonoprotective action of Zataria multiflora ethanolic extract on cyclophosphamide-induced oxidative lung toxicity in mice
Anti-leukemic effect of Zataria multiflora extract in combination with doxorubicin to combat acute lymphoblastic leukemia cells.
[198,199]
2-[(E)-3-naphtalen-2-yl-but-2-enoylamino]-benzoic acid (BIBR1532)Genes 14 00691 i002Monotherapy and combined with doxorubicinBIBR1532 exerts a series of anti-cancer activities linked to the inhibition of the canonical telomerase pathway and the TERT extra-telomeric functions in feline oral squamous cell carcinoma.
BIBR1532 exhibits a selective cytotoxicity against primary leukemia cells from acute myeloid leukemia and chronic lymphocytic leukemia patients.
Telomerase inhibition by BIBR1532 causes rapid cell death in pre-B-acute lymphoblastic leukemia cells
BIBR1532 exerted potent cytotoxic effects on a panel of human cancer cells in a dose-dependent manner in leukemic cells which were more sensitive to the inhibitor
BIBR 1532, exerts a direct short-term growth suppressive effect in a concentration-dependent manner possibly through the downregulation of c-Myc and hTERT expression
[46,200,201,202,203]
lipid-conjugated N30-P50 thiophosphoramidate GRN163L (Imetelstat)Genes 14 00691 i003MonotherapyImetelstat induces leukemia stem cell death in pediatric acute myeloid leukemia.
The telomerase antagonist imetelstat efficiently targets glioblastoma tumor-initiating cells leading to decreased proliferation and tumor growth
The inhibition of telomerase with imetelstat ex vivo led to significant dose-dependent apoptosis of B-ALL cells. Thus, imeteostat can be usefull in the standard treatment of B-ALL
[45,204,205]
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

da Mota, T.H.A.; Camargo, R.; Biojone, E.R.; Guimarães, A.F.R.; Pittella-Silva, F.; de Oliveira, D.M. The Relevance of Telomerase and Telomere-Associated Proteins in B-Acute Lymphoblastic Leukemia. Genes 2023, 14, 691. https://doi.org/10.3390/genes14030691

AMA Style

da Mota THA, Camargo R, Biojone ER, Guimarães AFR, Pittella-Silva F, de Oliveira DM. The Relevance of Telomerase and Telomere-Associated Proteins in B-Acute Lymphoblastic Leukemia. Genes. 2023; 14(3):691. https://doi.org/10.3390/genes14030691

Chicago/Turabian Style

da Mota, Tales Henrique Andrade, Ricardo Camargo, Estefânia Rodrigues Biojone, Ana Flávia Reis Guimarães, Fabio Pittella-Silva, and Diêgo Madureira de Oliveira. 2023. "The Relevance of Telomerase and Telomere-Associated Proteins in B-Acute Lymphoblastic Leukemia" Genes 14, no. 3: 691. https://doi.org/10.3390/genes14030691

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