SciELO - Scientific Electronic Library Online

 
 número37Simulación de lapercolación eléctrica de nanotubos de carbono a través de una matriz de polimetilmetacrilato: análisis bidimensional y tridimensional usando elefecto de latortuosidad de los nanoaditivosFrecuencia de enterobacterias productoras de carbapenemasas en hisopados anales de adultos en una unidad de cuidados críticos índice de autoresíndice de materiabúsqueda de artículos
Home Pagelista alfabética de revistas  

Servicios Personalizados

Revista

Articulo

Indicadores

  • No hay articulos citadosCitado por SciELO

Links relacionados

  • No hay articulos similaresSimilares en SciELO

Compartir


Revista de Ciencia y Tecnología

versión On-line ISSN 1851-7587

Rev. cienc. tecnol.  no.37 Posadas jun. 2022

http://dx.doi.org/10.36995/j.recyt.2022.37.009 

Articulos

Genetic and epi-genetic alterations of the PTEN gene: key in the relationship between type 2 diabetes and cancer

Alteraciones genéticas y epi-genéticas del gen PTEN: clave en la relación entre diabetes mellitus tipo 2 y cáncer

Alan Rolando Ayala Schimpf1  2 

María Mercedes Formichela4  5 

María Betania Mascheroni5  6 

Donovan Rivero1  2 

Marcela Luján Chamorro5 

Valeria Portillo6 

Pedro Darío Zapata1  2 

Cristian Alberto Ferri1  3  * 

1 Universidad Nacional de Misiones. Facultad de Ciencias Exactas Químicas y Naturales. Instituto de Biotecnología Misiones “Dra. María Ebe Reca” (INBIOMIS). Laboratorio de Biotecnología Molecular (BIOTECMOL). Misiones, Argentina.

2 CONICET. Buenos Aires, Argentina.

3 Instituto Universitario de Ciencias de la Salud, Fundación Héctor A. Barceló, Facultad de Medicina, Sede Santo Tomé, Centeno 710. Corrientes, Argentina.

4 Cátedra de Bioquímica Clínica II. Módulo de Farmacia y Bioquímica, Facultad de Ciencias Exactas Químicas y Naturales, Universidad Nacional de Misiones. Av. Mariano Moreno 1375 (3300) - Posadas, Misiones, Argentina.

5 Laboratorio CEBAC, Cede de la Clínica Nosiglia, Posadas, Misiones, Argentina.

6 Centro de Medicina Preventiva (PREDIGMA), Posadas, Misiones, Argentina.

Abstract

Cancer pathogenesis has been associated with genetic and epigenetic alterations that may lead to the inactivation or decrease of tumor-suppressor genes, such as the PTEN gene. In particular, alterations in the PI3K/PTEN signaling pathways are related to abnormalities associated with diabetes, metabolic syndrome, obesity and cardiovascular diseases. The aim of the study was to evaluate whether the expression levels of the PTEN gene, the 32-bp deletion in the PTEN gene, and the methylation in the PTEN promoter are associated with Type 2 Diabetes Mellitus(T2DM) and cancer development. Peripheral blood samples were obtained from a total of 70 patients of both sexes: 40 patients with T2DM and 30 patients with T2DM and cancer(T2DM+C). In this a novel report that assesses the upregulation of PTEN in T2DM, we observed that PTEN expression is higher in T2DM patients than in healthy individuals and markedly lower in T2DM patients with cancer than in T2DM patients without cancer. Hypermethylation of the PTEN promoter is more frequent in T2DM patients with cancer. Genetic and epigenetic alterations of the PTEN gene may be a key in the relationship between T2DM and cancer. PTEN expression may serve as a potential candidate biomarker for cancer development in T2DM patients.

Keywords: PTEN expression; Type 2 Diabetes Mellitus; Cancer; Hypermethylation

Resumen

La patogenia del cáncer ha sido asociada con alteraciones genéticas y epigenéticas que pueden llevar a la inactivación o subexpresión de genes supresores de tumores, como el gen PTEN. Las alteraciones en las vías de señalización de PI3K/PTEN están relacionadas con anomalías asociadas a diabetes, síndrome metabólico, obesidad y enfermedades cardiovasculares. El objetivo del estudio fue evaluar, si la expresión alterada (subexpresión/sobreexpresión) del gen PTEN, la deleción de 32 pb y la metilación del promotor, se asocian con diabetes mellitus tipo 2 (DMT2) y el desarrollo de cáncer. Las muestras de sangre periféricas se obtuvieron de un total de 70 pacientes de ambos sexos: 40 pacientes con DMT2 y 30 pacientes con DMT2 y cáncer (DMT2+C). En este trabajo novedoso, se reporta la sobreexpresión de PTEN en DMT2. Observamos que la expresión de PTEN es más alta en pacientes con DMT2 que en individuos sanos y marcadamente menor en pacientes con DMT2+C, en comparación con los pacientes diabéticos sin neoplasias. La hipermetilación del promotor de PTEN es más frecuente en pacientes con DMT2 y cancer. Las alteraciones genéticas y epigenéticas en el gen PTEN podría ser clave en la relación existente entre diabetes y el desarrollo de cancer. La expresión de PTEN puede ser un potencial biomarcador en el desarrollo cancer en pacientes con DMT2.

Palabras clave: Expresión de PTEN; Diabetes Mellitus Tipo 2; Cáncer; Hipermetilación

Introduction

Cancer pathogenesis has been associated with genetic and epigenetic alterations that may lead to the inactivation or decrease of tumor-suppressor genes 1. Among these, the phosphatase and tensin homolog (PTEN) gene is frequently deleted or inactivated in various tumors, suggesting that the loss of a single PTEN allele is sufficient to drive tumorigenesis. It has been reported that minimal changes in the expression level of PTEN can have profound effects on tumor susceptibility 4. In addition, mechanisms such as methylation or mutations in the gene promoter may lead to a loss of PTEN function. This loss of PTEN function is involved in the development of cancer and other human diseases 7-13. In addition, polymorphisms in the PTEN gene have been associated with Type 2 Diabetes Mellitus (T2DM)14-18.

T2DM is closely related to obesity and both are characterized by insulin resistance 19, generally defined as a reduction in the ability of insulin to exert its metabolic effects on tissues 20. Although the etiology of T2DM can be very different, both have been related to alterations in insulin secretion, in the sensitivity to the action of insulin, or both 21-23. Since insulin is also a powerful promoter of cell growth, it has been involved in the development of some types of cancer.. In fact, a strong relation between T2DM and the risk of developing certain types of cancer has been reported 25-27.

Insulin action activates two main signal transduction pathways: the phosphatidylinositol-3-kinase (PI3K) pathway and the mitogen-activated protein kinase (MAPK) pathway. Both pathways in turn regulate most insulin actions associated with the regulation of energy metabolism and gene expression28-30. In muscle tissue and adipocytes, the metabolic actions induced by insulin, including GLUT4 translocation, glucose uptake and glycogen synthesis, in turn depend on the activation of another pathway, the PI3K/Akt pathway. In addition, it has been demonstrated that abnormalities associated with diabetes, metabolic syndrome, obesity, hypertension and cardiovascular diseases are related to the PI3K/PTEN signaling pathway.

Thus, since T2DM and cancer are connected and PTEN may be a crucial gene related to both diseases, we consider it is essential to evaluate the PTEN expression pattern in T2DM patients and T2DM patients with cancer. Based on this, the aim of the present study was to evaluate PTEN expression, PTEN methylation and the frequent 32-bp deletion in the PTEN gene (known as rs34421660 genetic variant) and their relationship with T2DM and cancer development.

Materials and methods

Patients

To carry out this work, we have performed a case control study. All the participants provided an informed consent and their anonymity was preserved. The study was approved by the Ethics Committee of Dr. Fernando Barreyro Hospital, Posadas, Misiones, Argentina. A total of 135 samples were analyzed. Peripheral blood samples were obtained from 70 patients of both sexes, aged between 17 and 83 years old: 40 patients with T2DM and 30 patients with T2DM and cancer (T2DM+C). All samples were obtained at CEBAC laboratories of the Nosiglia Clinic, Posadas, Misiones, Argentina. Control samples were collected from 65 healthy individuals of both sexes, aged between 18 and 80 years old, all from Posadas.

Sample preparation

Total RNA and DNA were extracted from total leukocyte cells from peripheral using Trireagent® following the manufacturer’s protocol. First-strand cDNA synthesis was performed with random hexamer primers and M-MLV reverse transcriptase®.

Polymorphism detection

32-base-pair (bp) deletion polymorphism (rs34421660) was detected by conventional PCR, using Taq DNA polymerase 500U InBio Highway®.We obtain a fragment of 209 bp or 241 bp depending if the deletion was present or not. The primers used have been previously published 36 (Table 1). The reactions were carried out in the thermocycler Bioer GenePro. The rs34421660 polymorphism was corroborated by direct sequencing, using the PCR product corresponding to homozygous cases, one with deletion and the other without it (Figure 1). The PCR product was electrophoresed on 2% agarose gel containing ethidium bromide, a band corresponding to the expected size was purified using GFX columns (GE Healthcare) and then sequenced.

PTEN promoter methylation

The DNA samples were treated with sodium bisulfite, while methylated C residues resisted the treatment. Stanganelli et al. 37 method was used for the bisulfite treatment of DNA with certain modifications. Briefly, 1 μg of genomic DNA was denatured with 3 M NaOH at 37°C for 20 min and modified with 4.5 M sodium bisulfite (Sigma, St. Louis, MO, USA) and 10 mM hydroquinone (Sigma, St. Louis, MO, USA) at 55°C for 5 h. After purification with the WizardTM DNA Clean-up System (Promega A7280, Madison, WI, USA), the DNA was desulfonated in 3 M NaOH. The subsequent PCR amplification was performed using primers designed previously for either methylated or unmethylated DNA 38 (Table 1). The forward and reverse primers used for the first stage allowed amplify a fragment of 529 pb amplicon; 5 μl of the first-stage PCR product was used as a template for the second PCR stage. In the second stage, if the amplification product was unmethylated, the amplicon obtained was 173 bp, whereas, if the product was methylated, the amplicon obtained was 155 bp. Due to the difficulty in the process of methylation and subsequent amplification, only 42 samples could be analyzed by methylation of the PTEN promoter.

Thermal profiles consisted of an initial denaturation step at 95°C for 5 min, followed by 35 cycles at 97 °C for 45s, 58 °C for 30s, and 72 °C for 60 s, with a final extension step of 10 min at 72 °C. Reaction products were visualized after electrophoresis on 2% agarose gels (Tris-acetate EDTA

buffer, pH 8) containing ethidium bromide.

PTEN expression

To observe PTEN gene expression, Quantitative PCR was carried out using BIOER -Line Gene K Real Time, FDQ-48A (A4), as follows: 95 °C for 3 min, 50°C for 1 min, followed by 40 cycles to 95°C for 15 s and 60 °C for 1 min. The melting curve analysis and gel electrophoresis experiments were used to ensure that only one product of the expected size was amplified. Non-template controls were included in each run. Expression values were obtained from the threshold cycle (Ct value), using serial dilutions of the K562 cell line as standard curve.

The expression of the PTEN gene was normalized by subtracting the Ct value of the housekeeping gene (β-actin) from the Ct value of the target gene (ΔCt). The relative copy number of PTEN mRNA was calculated according to 2−ΔCt, and then expressed as a percentage. PTEN expression was measured in duplicate for each sample. The primers (table 1) and conditions have been previously published 39.

Statistical analysis

The association between the rs34421660 polymorphism and diabetes and cancer was analyzed by univariate Fisher's exact test and multivariable logistic regression analysis. The odds ratios (ORs) and their corresponding 95% confidence intervals (CIs) were calculated. The ORs were analyzed with the use of the SPSS software (version 15.0) (IBM, SPSS Inc., Chicago, USA), and Hardy-Weinberg equilibrium (HWE) was tested using a goodness-of-fit Chi-square test. Standard genetic models (additive, recessive, co-dominant and dominant) for disease penetrance were evaluated. Differences in expression levels were analyzed using one-way analysis of variance (ANOVA) and an unpaired t test to compare differences between groups. The statistical analyses were performed using GraphPad Prism5.0. All statistical tests were two-sided and values of p≤0.05 were considered statistically significant. To determine a value allowing the identification of cases with increased PTEN expression with respect to the healthy individuals (HI), a cut-off value from the healthy group was calculated as follows: mean + 3 standard deviation (SD) 39.

Results

Case-control study

Out of the 135 individuals incorporated in this study, 70 had T2DM and 30 of these individuals were also diagnosed with cancer (testicle, breast, colon, lung, prostate, kidney and pancreas). The distribution regarding gender and age is shown in Table 2.

The rs34421660 genotype frequencies were also analyzed comparing patients and controls according to DM2, DM2+C and the Control Group (Table 3). Genotype and allele frequency distributions for rs34421660 in cases and controls are shown in Table 4.

No deviation from HWE was found either for controls or patients (p=0.38 and p=0.45, respectively).

Table 1: Sequences of all PCR primers used in this study. 

Table 2: Patients: distribution by gender and age. 

Table 3: Distribution of the rs34421660 variant in T2DM, T2DM+C and Control Group. 

Table 4: Association study between the rs34421660 polymorphism and diabetes risk. 

Detection of the rs34421660 polymorphism (32-bp deletion)

After PCR amplification, all the samples were cataloged based on whether the rs34421660 polymorphism was present or absent considering gender and condition (patients or controls). The distribution of rs34421660 by condition is shown in Table 3.

The rs34421660 polymorphism seemed not to be associated with the development of diabetes [OR: 1.825 (0.804-4.140)] or cancer [OR: 1.64 (0.599-4.494)] and not involved in different levels of PTEN expression (p=0.45).

PTEN expression

The ANOVA (α=0.05) showed statistical differences in the PTEN expression levels (p<0.0001). Statistical parameter values of PTEN expression in the populations analyzed were as follows: statistical mean (+ HI: 3.69; +T2DM: 24.25; + T2DM+C: 7.74); SD σ (σ HI: 4.58; σ T2DM: 13.11; σ T2DM+C: 7.08); and standard error (SE) (SE HI: 0.57; SE T2DM: 2.62; SE T2DM+C: 1.67). The t test analysis was performed with significant level of α=0.05 and patients were compared between them and with HI. PTEN expression was statistically higher in T2DM patients than in HI (p<0.0001) and T2DM+C patients (p<0.0001). Besides, PTEN expression in T2DM+C patients was statistically different from that in HI (p= 0.0045) (Figure 2).

To determine a value that allow the identification of cases with increased PTEN expression with respect to HI, a cut-off value from the HI group was calculated as follows: mean + 3SD: 3.69 + (3 × 4.58) = 17.43. Unlike that observed in the other groups, 68% of T2DM patients presented an overexpression of the PTEN gene (Figure 3).

Methylation of the PTEN promoter

The methylation status of the PTEN promoter was analyzed in 42 cases of the three groups (Table 5). T2DM+C patients showed higher frequency of methylation than T2DM patients and HI (p=0.0215).

The first PCR stage, which produced a 529-bp amplicon, was used as a template for the second PCR stage, and the second stage was performed as previously described. In the second stage, the unmethylated amplification product was a 173-bp fragment, whereas the methylated amplification product was a 155-bp fragment.

Table 5: Methylation status of the PTEN gene promoter. 

Discussion

PTEN is a protein phosphatase with dual activity, both involved in the insulin signaling pathway and working as a tumor suppressor 42. The metabolic action of insulin depends on the PI3K signaling pathways, and some defects in these pathways have been reported in T2DM patients. In addition, PTEN can negatively regulate the positive actions of PI3K through the regulation of PIP3 levels. Thus, this negative regulation could "improve" the insulin signaling. Although the PTEN tumor-suppressor functions have already been widely demonstrated, the physiological role of PTEN in glucose metabolism is largely unknown.

Currently few studies are focusing on PTEN as a possible mediator by way of suppressor tumor, as well as negative regulator at the front desk of the insulin at the cellular level and its involvement in the development of cancer in individuals diagnosed with T2DM.

The aim of this work was to characterize the 32-bp deletion known as rs34421660 polymorphism in the PTEN gene and its association with the susceptibility to develop cancer in patients with T2DM. Besides, the levels of gene expression were assessed in patients whose PI3K pathway could be altered due to T2DM, and compared with respect to the expression levels in T2DM patients with cancer, considering PTEN functions as a tumor suppressor.

Polymorphisms can either be silent (and thus have no direct functional repercussions) or provide advantages or disadvantages against certain diseases in individuals carrying. Researches aimed to identify frequent genetic polymorphisms that confer susceptibility to suffer a particular disease are becoming more frequent. If these polymorphisms are present in the promoter region, they might derive in an altered gene expression. Similarly, when the changes occur in intronic regions, they could modulate the gene expression. In this context, specific molecular studies can demonstrate whether polymorphisms are functional, and epidemiological studies are fundamental to assess whether their presence is a risk factor for population health 49. In this study, no statistically significant association was observed between the rs34421660 polymorphism and T2DM and/or cancer development in T2DM patients. The role of PTEN as a tumor suppressor and its expression levels are regulated by several and complex mechanisms 41. The physiological action of PTEN is to block PI3K signaling by PIP3 dephosphorylation, avoiding related effects by PIP3 increase, as AKT signaling deregulation. An increase in AKT activity leads to amplified cell survival, growth and proliferation 51. It has been reported that patients diagnosed with many types of neoplasms have a lower expression of PTEN than those without neoplasms. These investigations are based on the development of cancer as the final result of the deregulation of the cell cycle and apoptosis inhibition. PTEN modulates the metabolic actions in the cell by regulating the pathway downstream PI3K, where, in addition to reducing the levels of PIP3, it can also dephosphorylate the insulin receptor substrate, altering the signaling of the insulin PI3K/AKT pathway through these two mechanisms 54. An increase in PTEN expression, and therefore in its activity, should lead to the same impact on the signaling of insulin as a decrease in the activity of the PI3K pathway 55.

In this study, the expression of PTEN presented a marked variation in the T2DM population with respect to healthy individuals. Through the cut-off value, calculated from the PTEN expression in healthy individuals, we identified that 68% of the T2DM patients studied had PTEN overexpression. A recent study related to the expression of this gene and its association with T2DM in a population of a region of Western China has recently found that this expression would be related to severe insulin resistance 6. In addition, PTEN is involved in the control of muscle protein metabolism, presenting a fluctuating behavior in catabolic conditions, and it has been shown that high levels of PTEN expression contribute to the degradation of muscle proteins, a characteristic phenomenon in T2DM 56. The T2DM mechanism that would lead to muscle atrophy is related to the reduction of protein synthesis. Proteolysis of the muscle in catabolic conditions is linked to insulin resistance and specifically to the defects in the substrate of the insulin receptor associated with the PI3K/AKT pathway. Experimental evidence has shown that PTEN levels vary with models of defects in insulin signaling, and that PTEN suppression produces a marked improvement of glucose concentrations in blood and of the insulin sensitivity in diabetic mice. On the other hand, overexpression of PTEN inhibits glucose uptake and GLUT4 translocation 57 and a high-fat diet-induced insulin resistance increases the activity of PTEN 58. The PTEN overexpression in patients with T2DM observed in this study is consistent with previous experimental evidence suggesting that PTEN is transcriptionally regulated in insulin chronic deficiency/resistance in T2DM. The mechanism of increased PTEN expression observed in T2DM would mean an increase in PTEN mRNA. The activity and expression of PTEN can be increased by the incubation of endothelial cells with free fatty acids. In this case, PTEN overexpression was related to the stimulation of p38 MAPK 59. Findings in muscle cells in culture have confirmed not only that palmitic acid increases p38 activation and PTEN expression, but also that p38 inhibition suppresses the increase in palmitic acid in PTEN mRNA and the expression of induced to protein. These results suggest that the transcription and expression of PTEN are related to the activation of p38, which could partly explain the high expression of PTEN in the T2DM.

In many tumors, the PTEN gene is mutated or deleted 42. Although in the present study we expected that T2DM patients with cancer would show decreased PTEN expression levels, we found no statistically significant difference with respect to those of the healthy individuals studied. Due to the great overexpression of PTEN observed in T2DM patients and considering that the diagnosis of cancer in the T2DM+C patients was subsequent to the T2DM condition, it is necessary to compare these two populations together. Although there was statistical difference selection to compare the levels of PTEN in T2DM patients with cancer and healthy individuals, the levels of expression in T2DM patients with cancer were markedly lower (p>0.0001) than those in T2DM patients without cancer. This may allow us to infer that, during T2DM, given the need to maintain over-expressed cellular homeostasis values of PTEN, variations in the levels of expression could lead to a deregulation of the "balance" reached in signaling pathways and promote the development of neoplasms. To prove this, it would be necessary to perform a prospective study in T2DM patients that develop any neoplasia over time and show decreased expression of PTEN. In addition to mutations in the PTEN gene, gene silencing through the hypermethylation of its promoter has been suggested as a potential mechanism that contributes to the down-regulation of the PI3K pathway by PTEN, which, at the same time, is positively associated with an increased risk to several types of cancer. This relationship between aberrant methylation and cancer has become evident by the fact that the absence of methylation leads to the activation of oncogenes and the presence of methylation annuls the protective action of tumor-suppressor genes. In the present study, we evaluated the methylation profile of the PTEN promoter in the different populations and established its association with the low expression of PTEN in T2DM patients with cancer. The inactivation of tumor-suppressor genes contributes to carcinogenesis by conferring certain advantages to indefinite cell growth, which is consistent with the partial results here obtained.

Conclusion

The rs34421660 polymorphism of PTEN in intron 2 was present in 44% of the healthy individuals studied. This polymorphism seems not to be associated with the development of T2DM or cancer as well as not involved in different levels of PTEN expression. However, PTEN expression levels are higher in T2DM patients than in healthy individuals and markedly lower in T2DM patients with cancer than in T2DM patients without cancer. The hypermethylated PTEN promoter is more frequent in T2DM patients with cancer. Genetic and epigenetic mechanisms of the PTEN gene may be a key in the relationship between T2DM and cancer. PTEN expression may serve as a potential candidate biomarker for cancer development in T2DM patients.

Legend to Figures

Figure 1: A: Representative polymerase chain reaction (PCR) products resolved by agarose gel electrophoresis to detect the presence or absence of the rs34421660 polymorphism (32-bp deletion) of the PTEN gene. M, DNA marker. Lane 1, heterozygous 32-bp deletion; lane 2: homozygous 32-bp deletion; lane 3: homozygous non-32-bp deletion. B: Genotype patterns for 32-bp homozygous deletion 

Figure 2: Percentage of the mean of PTEN expression. PTEN expression was statistically higher in T2DM patients than in healthy individuals (p<0.0001) and T2DM patients with cancer (p<0.0001). Furthermore, a statistically significant difference was observed between T2DM+C patients and healthy individuals (p= 0.0045). HI: Healthy individuals, T2DM: patients with Diabetes Mellitus Type 2, T2DM+C: patients with Type 2 Diabetes Mellitus and cancer. 

Figure 3: Frequency of cases with PTEN expression >17.43. Percentage calculated taking into account the cut-off value obtained from HI (+ 3SD = 17.43). High expression levels of PTEN among T2DM compared with T2DM+C and HI. Dark gray column: PTEN expression >17.43. Gray column: PTEN expression <17.43. HI: Healthy individuals, T2DM: patients with Type 2 Diabetes Mellitus, T2DM+C: patients with Type 2 Diabetes Mellitus and cancer. 

Acknowledgments

This study was supported in part by grant from the National Agency of Scientific and Technical Promotion (SECyT-PICT).

Thanks to Professor Lic. Ma. Victoria Gonzalez Eusevi for editing and proofreading the article.

References

1. Waniczek D, Snietura M, Kopec A, et al. A novel quantitative method of pten expression assessment in tumor tissue. Journal of biological regulators and homeostatic agents 2016;30(1): 79-90. [ Links ]

2. Berger AH, Knudson AG, Pandolfi PP. A continuum model for tumour suppression. Nature 2011;476(7359): 163. [ Links ]

3. Yan Y, Huang H. Interplay Among PI3K/AKT, PTEN/FOXO and AR Signaling in Prostate Cancer. Prostate Cancer: Springer, 2019:319-31. [ Links ]

4. Hopkins B, Hodakoski C, Barrows D, Mense SM, Parsons RE. PTEN function: the long and the short of it. Trends in biochemical sciences 2014;39(4): 183-90. [ Links ]

5. Yin L, Cai W-J, Liu C-X, et al. Analysis of PTEN methylation patterns in soft tissue sarcomas by MassARRAY spectrometry. PloS one 2013;8(5): e62971. [ Links ]

6. Yin L, Cai W J, Chang X Y, et al. Analysis of PTEN expression and promoter methylation in Uyghur patients with mild type 2 diabetes mellitus. Medicine 2018;97(49). [ Links ]

7. Pulido R. PTEN: a yin-yang master regulator protein in health and disease. Methods 2015;77-78: 3-10. [ Links ]

8. Pulido R, van Huijsduijnen RH. Protein tyrosine phosphatases: dual‐specificity phosphatases in health and disease. The FEBS journal 2008;275(5): 848-66. [ Links ]

9. Osei Amponsa V, Buckwalter JM, Shuman L, Zheng Z, Yamashita H, Walter V, et al. Hypermethylation of FOXA1 and allelic loss of PTEN drive squamous differentiation and promote heterogeneity in bladder cancer. Oncogene 2020; 39(6): 1302-1317. [ Links ]

10. Bell CG, Finer S, Lindgren CM, et al. Integrated genetic and epigenetic analysis identifies haplotype-specific methylation in the FTO type 2 diabetes and obesity susceptibility locus. PLoS One 2010;5(11): e14040. [ Links ]

11. Dayeh T, Volkov P, Salö S, et al. Genome-wide DNA methylation analysis of human pancreatic islets from type 2 diabetic and non-diabetic donors identifies candidate genes that influence insulin secretion. PLoS genetics 2014;10(3): e1004160. [ Links ]

12. Alfieri R, Giovannetti E, Bonelli M, Cavazzoni A. New treatment opportunities in phosphatase and tensin homolog (PTEN)-Deficient tumors: focus on PTEN/focal adhesion kinase pathway. Frontiers in oncology 2017;7: 170. [ Links ]

13. Russo A, Czarnecki AA, Dean M, et al. PTEN loss in the fallopian tube induces hyperplasia and ovarian tumor formation. Oncogene 2018;37(15): 1976. [ Links ]

14. Khokhar M, Roy D, Modi A, et al. Perspectives on the role of PTEN in diabetic nephropathy: an update. Critical reviews in clinical laboratory sciences 2020; 57(7): 470-483. [ Links ]

15. Khokhar M, Roy D, Modi A., Agarwal R, et al. Perspectives on the role of PTEN in diabetic nephropathy: an update 2020. Critical reviews in clinical laboratory sciences, 57(7), 470-483. [ Links ]

16. Khorami SAH, Abd Mutalib MS, Shiraz MF, et al. 2020. Genetic determinants of obesity heterogeneity in type II diabetes 2020. Nutrition & Metabolism, 17(1): 1-9. [ Links ]

17. Baroudi T, Bouhaha R, Moran-Moguel C, et al. Association of the insertion/deletion polymorphism of the angiotensin-converting enzyme gene with type 2 diabetes in two ethnic groups of Jerba Island in Tunisia. Journal of the Renin-Angiotensin-Aldosterone System 2009;10(1): 35-40. [ Links ]

18. Chen C Y, Chen J, He L, Stiles BL. PTEN: tumor suppressor and metabolic regulator. Frontiers in endocrinology 2018;9. [ Links ]

19. Peng J, He L. IRS posttranslational modifications in regulating insulin signaling. Journal of molecular endocrinology 2018;60(1): R1-R8. [ Links ]

20. Smith U. PTEN-linking metabolism, cell growth, and cancer: Mass Medical Soc, 2012. [ Links ]

21. Chiasson J L. Prevention of Type 2 diabetes: fact or fiction? Expert Opin Pharmacother 2007;8(18): 3147-58. [ Links ]

22. Gower BA, Goss AM. The sliding set-point: how insulin and diet interact to explain the obesity epidemic (and how to fix it). Current Opinion in Endocrinology, Diabetes and Obesity 2018;25(5): 303-09. [ Links ]

23. Akash MSH, Rehman K, Liaqat A. Tumor necrosis factor‐alpha: role in development of insulin resistance and pathogenesis of type 2 diabetes mellitus. Journal of cellular biochemistry 2018;119(1): 105-10. [ Links ]

24. Giovannucci E, Harlan DM, Archer MC, et al. Diabetes and cancer: a consensus report. CA: a cancer journal for clinicians 2010;60(4): 207-21. [ Links ]

25. Smith U, Gale E. Cancer and diabetes: are we ready for prime time? Diabetologia 2010;53(8): 1541-44. [ Links ]

26. Pal A, Barber TM, Van de Bunt M, et al. PTEN mutations as a cause of constitutive insulin sensitivity and obesity. New England Journal of Medicine 2012;367(11): 1002-11. [ Links ]

27. Pal A, Barber TM, Van de Bunt M, et al. PTEN mutations as a cause of constitutive insulin sensitivity and obesity in human beings. The Lancet 2013;381: S13. [ Links ]

28. Youngren J. Regulation of insulin receptor function. Cellular and Molecular Life Sciences 2007;64(7-8): 873. [ Links ]

29. Hopkins BD, Goncalves MD., & Cantley LC. Insulin-PI3K signalling: an evolutionarily insulated metabolic driver of cancer. Nature Reviews Endocrinology 2020; 16(5): 276-283. [ Links ]

30. Lazar DF, Saltiel AR. Lipid phosphatases as drug discovery targets for type 2 diabetes. Nature Reviews Drug Discovery 2006;5(4): 333. [ Links ]

31. Shahbazi R, Cheraghpour M, Homayounfar R, Nazari M, Nasrollahzadeh J, Davoodi S. Hesperidin inhibits insulin-induced phosphoinositide 3-kinase/Akt activation in human pre-B cell line NALM-6. Journal of cancer research and therapeutics 2018;14(3): 503-08. [ Links ]

32. Ravi Y, Selvendiran K, Meduru S, et al. Dysregulation of PTEN in cardiopulmonary vascular remodeling induced by pulmonary hypertension. Cell biochemistry and biophysics 2013;67(2): 363-72. [ Links ]

33. Mitchell F. PTEN mutations increase insulin sensitivity and obesity. Nature reviews Endocrinology 2012;8(12): 698-99. [ Links ]

34. Johnson DL, Stiles BL. Maf1, a new PTEN target linking RNA and lipid metabolism. Trends in Endocrinology & Metabolism 2016;27(10): 742-50. [ Links ]

35. Boosani CS, Agrawal DK. PTEN modulators: a patent review. Expert opinion on therapeutic patents 2013;23(5): 569-80. [ Links ]

36. Hashemi M, Rezaei H, Eskandari-Nasab E, Kaykhaei M-A, Taheri M. Association of promoter methylation and 32-bp deletion of the PTEN gene with susceptibility to metabolic syndrome. Mol Med Rep 2013;7(1): 342-46. [ Links ]

37. Stanganelli C, Arbelbide J, Fantl DB, Corrado C, Slavutsky I. DNA methylation analysis of tumor suppressor genes in monoclonal gammopathy of undetermined significance. Ann Hematol 2010;89(2): 191. [ Links ]

38. Goel A, Arnold CN, Niedzwiecki D, et al. Frequent inactivation of PTEN by promoter hypermethylation in microsatellite instability-high sporadic colorectal cancers. Cancer Res 2004;64(9): 3014-21. [ Links ]

39. Ferri C, Bianchini M, Bengió R, Larripa I. Expression of LYN and PTEN genes in chronic myeloid leukemia and their importance in therapeutic strategy. Blood Cells, Molecules, and Diseases 2014;52(2-3): 121-25. [ Links ]

40. Goel A, Arnold CN, Niedzwiecki D, et al. Frequent inactivation of PTEN by promoter hypermethylation in microsatellite instability-high sporadic colorectal cancers. Cancer research 2004;64(9): 3014-21. [ Links ]

41. Hashemi M, Rezaei H, Eskandari Nasab E, Kaykhaei M A, Taheri M. Association of promoter methylation and 32-bp deletion of the PTEN gene with susceptibility to metabolic syndrome. Molecular medicine reports 2013;7(1): 342-46. [ Links ]

42. Hopkins BD, Hodakoski C, Barrows D, Mense SM, Parsons RE. PTEN function: the long and the short of it. Trends in biochemical sciences 2014;39(4): 183-90. [ Links ]

43. Taniguchi CM, Emanuelli B, Kahn CR. Critical nodes in signalling pathways: insights into insulin action. Nature reviews Molecular cell biology 2006;7(2): 85. [ Links ]

44. Butler M, McKay RA, Popoff IJ, et al. Specific inhibition of PTEN expression reverses hyperglycemia in diabetic mice. Diabetes 2002;51(4): 1028-34. [ Links ]

45. Guttmacher AE, Collins FS. Genomic medicine-a primer. New England Journal of Medicine 2002;347(19): 1512-20. [ Links ]

46. Sun L, Liu J, Yuan Q, Xing C, Yuan Y. Association between PTEN Gene IVS4 polymorphism and risk of cancer: a meta-analysis. PloS one 2014;9(6): e98851. [ Links ]

47. Porta M. The genome sequence is a jazz score. International Journal of Epidemiology 2003;32(1): 29-31. [ Links ]

48. Sun L, Wang Q, Gao X, Shi D, Mi S, Han Q. MicroRNA-454 functions as an oncogene by regulating PTEN in uveal melanoma. FEBS letters 2015;589(19): 2791-96. [ Links ]

49. Caporaso NE. Why have we failed to find the low penetrance genetic constituents of common cancers? Cancer Epidemiology and Prevention Biomarkers 2002;11(12): 1544-49. [ Links ]

50. Leslie NR, Yang X, Downes CP, Weijer CJ. PtdIns (3, 4, 5) P3-dependent and-independent roles for PTEN in the control of cell migration. Current Biology 2007;17(2): 115-25. [ Links ]

51. Di Cristofano A, Pandolfi PP. The multiple roles of PTEN in tumor suppression. Cell 2000;100(4): 387-90. [ Links ]

52. Hassan M, Watari H, AbuAlmaaty A, Ohba Y, Sakuragi N. Apoptosis and molecular targeting therapy in cancer. BioMed research international 2014;2014. [ Links ]

53. Bosch-Barrera J, Hernandez A, Abella L. Insulin and insulin-like growth factor pathway, a new targeted therapy in oncology. Anales del sistema sanitario de Navarra, 2009:413-21. [ Links ]

54. Gutiérrez-Rodelo C, Roura-Guiberna A, Olivares-Reyes JA. Molecular mechanisms of insulin resistance: An update. Gaceta medica de Mexico 2017;153(2): 214-28. [ Links ]

55. Dupont J, Renou JP, Shani M, Hennighausen L, LeRoith D. PTEN overexpression suppresses proliferation and differentiation and enhances apoptosis of the mouse mammary epithelium. The Journal of clinical investigation 2002;110(6): 815-25. [ Links ]

56. Hu Z, Lee IH, Wang X, et al. PTEN expression contributes to the regulation of muscle protein degradation in diabetes. Diabetes 2007;56(10): 2449-56. [ Links ]

57. Nakashima N, Sharma PM, Imamura T, Bookstein R, Olefsky JM. The tumor suppressor PTEN negatively regulates insulin signaling in 3T3-L1 adipocytes. Journal of Biological Chemistry 2000;275(17): 12889-95. [ Links ]

58. Lo Y, Tsao C, Liu I, Liou S, Cheng J. Increase of PTEN gene expression in insulin resistance. Hormone and Metabolic Research 2004;36(10): 662-66. [ Links ]

59. Wang XL, Zhang L, Youker K, et al. Free fatty acids inhibit insulin signaling-stimulated endothelial nitric oxide synthase activation through upregulating PTEN or inhibiting Akt kinase. Diabetes 2006;55(8): 2301-10. [ Links ]

60. Salmeron J, Hu FB, Manson JE, et al. Dietary fat intake and risk of type 2 diabetes in women. The American journal of clinical nutrition 2001;73(6): 1019-26. [ Links ]

Received: June 14, 2021; Accepted: October 15, 2021

Creative Commons License This is an open-access article distributed under the terms of the Creative Commons Attribution License