Skip to main content
Erschienen in: Cancer Cell International 1/2022

Open Access 01.12.2022 | Review

A review on the role of PTENP1 in human disorders with an especial focus on tumor suppressor role of this lncRNA

verfasst von: Soudeh Ghafouri-Fard, Tayyebeh Khoshbakht, Bashdar Mahmud Hussen, Mohammad Taheri, Nader Akbari Dilmaghani

Erschienen in: Cancer Cell International | Ausgabe 1/2022

Abstract

PTENP1 is a long non-coding RNA which has been regarded as a pseudogene of the PTEN tumor suppressor gene. However, it has been shown to be a biologically active transcript that can function as a competing endogenous RNA and enhance expression of PTEN protein. This lncRNA has two transcripts, namely PTENP1-202 and PTENP1-202 with sizes of 3996 and 1215 bps, respectively. PTENP1 acts as a sponge for some PETN-targeting miRNAs, such as miR-17, miR-20a, miR-19b, miR-106b, miR-200c, miR-193a-3p, miR-499-5p and miR-214. Besides, it can affect miR-20a/PDCD4, miR-27a-3p/EGR1, miR-17‐5p/SOCS6 and miR-19b/TSC1 axes. This long non-coding RNA participates in the pathoetiology of several types of cancers as well as non-malignant conditions such as alcohol-induced osteopenia, insulin resistance, osteoporosis, sepsis-associated cardiac dysfunction and spinal cord injury. In the current review, we elucidate the role of PTENP1 in human disorders, particularly malignant conditions based on evidence acquired from cell line assays, animal studies and investigations on human samples.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Long non-coding RNAs (lncRNAs) are a group of RNAs with sizes longer than 200 nucleotides, several shared features with mRNAs, the ability to regulate gene expression and lack of significant open reading frames. This novel group of epigenetic regulators mainly resides in the nucleus where they affect histone or DNA modification, chiefly methylation and acetylation [1]. Through influencing alternative splicing, cell differentiation, and cell cycle transition, lncRNAs contribute in the evolution of many diseases [24]. Moreover, lncRNAs can affect the organization and function of nuclear bodies, modify the stability and expression of cytoplasmic mRNAs and regulate activity of signaling pathways [5]. Functions and contribution of several lncRNAs in human diseases have been reviewed [68].
Phosphatase and Tensin Homolog Pseudogene 1 (PTENP1) is an example of lncRNAs which has been regarded as a pseudogene of the PTEN tumor suppressor gene. However, it has been shown to be a biologically active transcript that can function as a competing endogenous RNA (ceRNA) and enhance expression of PTEN protein [9]. In fact, PTENP1 exerts a growth-suppressive effect through obstructing the binding of miRNAs to the 3′ UTR of PTEN and protecting it from degradation [9].
The gene coding this lncRNA is located on chromosome 9: 33,673,504−33,677,499 reverse strand. This lncRNA has two transcripts, namely PTENP1-202 and PTENP1-202 with sizes of 3996 and 1215 bps, respectively (https://​asia.​ensembl.​org/​Homo_​sapiens/​Gene/​Summary?​g=​ENSG00000237984;r=​9:​33673504-33677499). In the current review, we elucidate the function of PTENP1 in human disorders, particularly malignant conditions based on evidence obtained from cell line assays, animal studies and investigations on human samples.

Cell line studies

An in vitro experiment in HL-60 promyeoloblastic cells infected with the pCDH1-PTENP1 vectors has shown up-regulation of both PTENP1 and PTEN mRNA levels. However, protein levels of PTEN have not been affected by this intervention. Authors have suggested that PTENP1 can affect PTEN expression at mRNA level [10].
In addition to hematopoietic cells, PTENP1 can affect malignant properties of cell lines originated from solid tumors. Normal cells can secret PTENP1 in their exosomes. Exosome-mediated transmission of this lncRNA to bladder cancer cells could inhibit the malignant features in these cells through induction of cell apoptosis and reduction of invasion and migration abilities of bladder cancer cells. Functionally, exosomal PTENP1 could increase PTEN expression through sponging miR-17 [11]. The PTENP1/miR-20a/PTEN molecular route has been shown to affect malignant behavior of bladder cancer cells. While up-regulation of miR-20a could promote proliferation and migration of T24 cells, PDCD4 over-expression could exert the opposite effects [12].
Expression levels of PTENP1 have also been assessed in breast cancer cells. PTENP1 has also been shown to influence proliferation, invasive properties and resistance of breast cancer cells to Adriamycin. These effects are most probably mediated through sponging miR-20a and further regulating expression of PTEN and activity of PI3K/AKT pathway [13]. Moreover, this lncRNA could affect breast cancer pathogenesis through modulation of miR-19b/PTEN axis [14]. PTENP1 could also suppress proliferation and migratory aptitude of breast cancer cells via decreasing expressions of cell cycle regulators cyclin A2 and CDK2 and regulating activity of AKT and MAPK pathways [15]. Finally, the sponging role of PTENP1 on miR-19b has been shown to be implicated in the suppression of proliferation and of breast cancer cells [16] (Fig. 1).
Similarly, PTENP1 could inhibit progression of cervical cancer through different mechanisms including suppression of miR-106b [17], miR-27a-3p [18] and miR-19b [19]. These miRNAs target PTEN, EGR1 and MTUS1, respectively (Fig. 2).
Figure 1. Summary of the role of PTENP1 in progression of cancers. PTENP1 can serve as molecular sponge for miR-19b, miR-20a and miR-17. Down-regulation of these miRNAs by PTENP1 affects proliferation, migration and invasiveness of cancer cells. Detailed information about the impact of this lncRNA on suppression of carcinogenesis is provided in Table 1.
Figure 2. Summary of the role of PTENP1 in progression of cancers. PTENP1 can serve as molecular sponge for miR-21, miR-10a-5p, miR-19b, miR-27a-3p, miR-193a-3p, miR-19b, miR-20a and miR-17. Down-regulation of these miRNAs by PTENP1 induces anti-tumor effects. Detailed information about the impact of this lncRNA on suppression of carcinogenesis is provided in Table 1.
Table 1
Role of PTENP1 in different cancers according to cell line studies 
Tumor
Interactions
Cell line
Function
References
Acute leukemia
PTEN
HL-60 cell line and 293T cells
↑↑ PTENP1: ↑ PTEN mRNA level without affecting PTEN protein levels and cell growth
[10]
Bladder cancer
miR-17/PTEN axis
EJ, J82, HEK 293 A
↑↑ PTENP1: ↓ proliferation, migration, invasion, colony formation, ↑ apoptosis
[11]
miR-20a/PDCD4 axis
Human bladder cancer cell lines J82 and T24, SV-HUC-1
↑↑ miR-20a (a target of PTENP1): ↑ proliferation and migration
[12]
Breast cancer
miR-20a/PTEN axis, PI3K/Akt signaling
MDA-MB-231, T-47D and MCF-7 , mammary epithelium MCF-10 A
↑↑ PTENP1: ↓ proliferation, migration, invasion, colony formation, viability
[13]
miR-19b/ PTEN axis, p53 and p-AKT
MCF-10 A,BT-20, MCF-7, MDA-MB-231 and T-47D
↑↑ PTENP1: ↓ proliferation, migration, invasion, ↑ apoptosis
↑ p53 and ↓p-AKT
[14]
AKT and MAPK signaling pathways
MCF7, 293T
↑↑ PTENP1: ↓ proliferation, migration, colony formation, cyclin A2 and CDK2, AKT and MAPK signaling pathways
[15]
miR-19b/ PTEN axis and PI3K/Akt Pathway
MCF10A, MCF-7 and MDA-MB-231
↑↑ PTENP1: ↓ proliferation, migration, invasion, colony formation, PDK-1, p-PI3K, PI3K, and p-Akt, ↑ apoptosis, PTEN
[16]
Cervical cancer
miR-106b/ PTEN axis
HeLa, SiHa, C33A, CasKi, H8
↑↑ PTENP1: ↓ proliferation, EMT process, ↑ apoptosis
[17]
miR-27a-3p/ EGR1 axis
C33A, HeLa, ME-180, SiHa, NC104
↑↑ PTENP1: ↓ proliferation, EMT process, motility, ↑ apoptosis
[18]
miR-19b/ MTUS1 axis
Human normal cervical epithelium cell (HCvEpC) and human CC cell lines, such as Caski, C33A, SiHa and HeLa cells
↑↑ PTENP1: ↓ proliferation and invasion
[19]
Endometrioid endometrial carcinoma
miR-200c/ PTEN axis and PI3K-AKT pathway
RL-952, Ishikawa, HEC-1B, and JEC
17β-estradiol (E2) treatment: ↑ proliferation, migration and invasion, miR-200c levels, phospho-PI3K-AKT pathway genes and ↓ PTEN level
∆ ERα: ↓ effects of E2 on miR-200c and PTEN
[20]
Esophageal carcinoma
miR-17‐5p/ SOCS6 axis, p‐STAT3‐HIF‐1α signal pathway
Eca109, TE-1, HEK‐293T, Het‐1 A
↑↑ PTENP1: ↓ proliferation, vitality, p-STAT3‐HIF‐1α signal pathway
[21]
Gastric cancer
miR-106b, miR-93 and PTEN
GES-1, gastric adenocarcinoma cell line AGS, SGC7901, MGC803 and BGC823
↑↑ PTENP1: ↓ cell growth, migration, and invasion, ↑ apoptosis
[22]
Glioma
p21 and p38 signaling pathway.
SHG44 and U251 human glioma cells
↑↑ PTENP1: ↓ proliferation, migration, and invasion, p38 MAPK signaling pathway, ↑ cell cycle arrest, p21 levels
[23]
miR-10a-5p/ PTEN axis
Glioma cell line U87
Co-Culture of hUC-MSCs-derived exosomes suppress the proliferation and stimulate the apoptosis of U87 Cells.
Exosomes-Mediated Transfer of LncRNA PTENP1 suppresses Cell Growth by Targeting MiR-10a-5p.
[24]
Head and neck squamous cell carcinoma
PTEN
WSU-HN4, HN6, HN13, HN30 and Cal27
↑↑ PTENP1: ↓ cell growth, migration, invasion, colony formation
[25]
Hepatocellular carcinoma
miR-21, TET1/2/3, PTEN
SNU-449, HepG2, Hep3B, Huh7
↑↑ miR-21: ↑ proliferation, invasion, ↓ apoptosis, expression of TET1/2/3, change in methylation and expression of PTENp1, ↓ PTENp1 and PTEN
[26]
miR-193a-3p/ PTEN axis
Sk-Hep-1 and SMMC-7721
↑↑ PTENP1: ↓ proliferation, migration, invasion, ↑ apoptosis
[27]
miR-17, miR-19b and miR-20a, PTEN, PHLPP, ULK1, ATG7 and p62, ↓ PI3K/AKT pathway
human hepatocytes (HH) and HCC
cell line Mahlavu
↑↑ PTENP1: ↓ proliferation, migration, invasion ↑ autophagy and apoptosis
[28]
Multiple myeloma
miR-19b/ TSC1 axis
OPM2 and KMS-11 cells
↑↑ PTENP1: ↓ miR-19b levels and ↑ proliferation
[29]
Oral squamous cell carcinoma
miR-21/ PTEN, AKT pathways
SCC-25, Cal-27, and HEK 293 cells and ca-8113,
↑↑ PTENP1: ↓ proliferation, ↑ cell cycle arrest
[30]
Renal cell carcinoma
miR21/ PTEN axis
Human renal cell carcinoma cell lines 786-O, ACHN, SN12PM6 and HK-2
↑↑ PTENP1: ↓ proliferation and cell growth, migration, invasion, metastasis, and ↑ sensitivity of ccRCC cells to cisplatin and gemcitabine
[31]
BC breast cancer, ccRCC clear-cell renal cell carcinoma, ∆ knock-down or deletion
PTENP1 can also affect pathoetiology of non-malignant conditions (Table 2). For instance, it can affect pathogenesis of alcohol-induced osteopenia. Ethanol stimulation has resulted in up-regulation of expression of PTEN and PTENP1 transcripts in a time-dependent mode, leading to up-regulation of PTEN protein levels. Moreover, ethanol could decrease PTEN phosphorylation, representing an upsurge in functional PTEN level. Up-regulation of PTEN could impair downstream Akt/GSK3β/β-catenin signals and osteogenic differentiation of bone mesenchymal stem cells [32]. Moreover, PTENP1 binding to miR-499-5p leads to deficiency in the insulin-signaling pathway, thus participating in insulin resistance [33]. Furthermore, up-regulation of PTENP1 or silencing of miR-214 could inhibit expressions of osteoclast markers and RANKL-induced osteoclast differentiation. These interventions could also inhibit phosphorylation of PI3K and AKT, nuclear transport of p65, destruction of IκBα and NFATc1 expression. On the other hand, PTENP1 silencing has enhanced osteoclast differentiation. Taken together, PTENP1 acts as a sponge for miR-214 to escalate expression of PTEN and suppress osteoclast differentiation. This mode of action attenuates osteoporosis through inhibition of PI3K/AKT/NF-κB signaling [34].
Table 2
Role of PTENP1 in different non-malignant conditions according to cell line studies
Disorders
Interaction
Cell line
Function
References
Alcohol-induced osteopenia
PTEN and Akt/GSK3β/β-catenin signaling
Human BMSCs (hBMSCs)
Ethanol treatment: ↑ PTEN and PTENP1 levels and ↓ Akt/GSK3β/β-catenin signaling
∆ PTEN: ↓ ethanol-induced
suppression of bone formation and antiosteogenic effect of ethanol
[32]
Insulin resistance
miR-499-5p/ PTEN axis
Murine liver cell line NCTC1469
↑↑ PTEN: ↓ Akt/GSK activation and glycogen synthesis
[33]
Osteoporosis
miR-214/ PTEN axis, 3 K/AKT/NF-kB signaling pathway
RAW 264.7 macrophages
↑↑ PTENP1: ↓ ANKL-
induced osteoclast differentiation BY inhibiting 3 K/AKT/NF-kB signaling pathway
[34]
Sepsis-associated cardiac dysfunction
miR-106b-5p
H9C2
Matrine administration: ↓ expression of PTENP1 and inflammation, ↑ H9C2 viability
[35]
Spinal cord injury
miR-21, miR-19b and PTEN
SH-SY5Y and U251 cells
∆ PTENP1: ↑ apoptosis, miR-21, miR-19b and ↓ cell viability
[36]

Animal studies

Impact of PTENP1 up-regulation and exosomal PTENP1 on growth of tumors has been investigated in vivo. Authors have injected EJ cells with PTENP1-expressing vectors as well as PTENP1-containing exosomes into nude mice. The results of conducted experiments have indicated that up-regulation of PTENP1 can decrease tumor weight and burden. Moreover, PTENP1-containing exosomes could attenuate tumor size and weight. Besides, over-expression of this lncRNA could reduce Ki67 expression in tumors [11]. Other studies in esophageal carcinoma, head and neck squamous cell carcinoma, hepatocellular cancer and oral squamous cell carcinoma have confirmed the impact of PTENP1 up-regulation on attenuation of tumor growth (Table 3). In animal models of renal cell carcinoma, up-regulation of this lncRNA has enhanced sensitivity to cisplatin and gemcitabine [31].
Animal models have also been used to evaluate the impact of PTENP1 in insulin resistance. An experiment in db/db mice and high fat diet-fed mice has shown up-regulation of PTENP1. Moreover, up-regulation of PTENP1 has led to impairment in activation of Akt/GSK and production of glycogen, while suppression of this lncRNA has enhanced activity of Akt/GSK and increased glycogen content [33]. In an in vivo study, it has shown that the effect of matrine on improvement of cardiac function and attenuation of the inflammatory responses is mediated through down-regulation of PTENP1 expression and up-regulation of miR-106b-5p levels [35].
Table 3
PTENP1 role in different disorders based on animal studies
Tumor/ disease type
Animal models
Results
References
Bladder cancer
5 week-old male nude mice injected with EJ cell lines
↑↑ PTENP1: ↓ tumor weight, tumor volume and tumor size
[11]
Esophageal carcinoma
4 week-old male nude mice injected with Eca109 cells transfected with PTENP1 3′UTR or NC
↑↑ PTENP1: ↓ tumorigenesis
[21]
Head and neck squamous cell carcinoma
4-week-old male nude mice
↑↑ PTENP1: ↓ tumorigenesis
[25]
Hepatocellular carcinoma
4 week-old BALB/c nude mouse
∆ miR-21: ↓ tumor growth and size, ↑ PTEN, PTENp1, TET1, TET2 and TET3
[26]
4 week-old male immune-deficient nude mice (BALB/c-nu)
↑↑ PTENP1: ↓ tumor weight and tumor volume
[27]
6-8-weeks-old BALB/c nude mic were injected with Mahlavu cells
↑↑ PTENP1: ↓ tumor growth, intratumoral cell proliferation, and angiogenesis, ↑ apoptosis, autophagy
[28]
Oral squamous cell carcinoma
5 -week-old female BALB/C nude mice
mice were injected with Tca-8113 cells transfected with LV-miR-21 plus LV-PTEN and LV-PTENp1
↑↑ PTENP1: ↓ tumorigenesis
[30]
Renal cell carcinoma
Nude mice were injected with ACHN cells transfected with vector control or PTENP1
↑↑ PTENP1: ↑ sensitivity of ccRCC cells to cisplatin and gemcitabine
[31]
Alcohol-induced osteopenia
40 8-week-old male specific SPF and SD rats
∆ PTEN: ↓ ethanol-induced osteopenia
[32]
Insulin resistance
5 db/db mice and 5 age-matched wild-type (WT) mice
↑↑ PTENP1: ↑ hepatic insulin resistance
[33]
Osteoporosis
8-week-old female C57BL/6 mice
Levels of PTENP1 and PTEN were down-regulated in CS-F- and
RANKL-induced bone marrow mononuclear cell.
[34]
Spinal cord injury
Rats in sham group and SCI, SCI + exosomes, and SCI + exosomes + PTENP1-shRNA groups
Treatment with exosomes + PTENP1-shRNA: ↓ PTEN expression
PTENP1 participates in the recovery of SCI through regulation of miR-19b and miR-21.
[36]
knock-down or deletion, ccRCC clear-cell renal cell carcinoma, SPF specific pathogen-free, SD Sprague–Dawley

Clinical studies

Expression of PTEN and PTENP1 mRNAs has been demonstrated to be lower in bone marrow samples of AML patients compared to healthy subjects. Moreover, expressions of these transcripts have been positively correlated. However, when AML patients have been classified based on the prognostic classification of 2011 NCCN, authors have detected no remarkable difference in the expression of PTENP1 among subgroups [10].
Expression of PTENP1 has also been shown to be diminished in bladder cancer tissues as well as exosomes extracted from plasma samples of these patients. In fact, this lncRNA has been found to be principally carried by exosomes. Exosomal levels of PTENP1 have the potential to discriminate bladder cancer patients from healthy subjects with area under receiver characteristic curve of 0.743. Thus, exosomal PTENP1 has been recommended as a putative marker for diagnostic purposes in bladder cancer [11]. In bladder cancer cells, PTENP1 target miR-20a has been shown to be up-regulated, while PDCD4 has been down-regulated [12].
In breast cancer, cervical cancer, head and neck squamous cell carcinoma, hepatocellular carcinoma and oral squamous cell carcinoma, down-regulation of PTENP1 has been linked with poor survival of patients (Table 4). Moreover, down-regulation of this lncRNA has been correlated with advanced histological grade and TNM stage, deep infiltration depth, and lymph node metastasis in cancer patients.
Association between a number of tag single nucleotide polymorphisms within PTENP1, including rs7853346 C > G, rs865005 C > T, and rs10971638 G > A and susceptibility to gastric cancer has been assessed in a Chinese population. Results have shown association between rs7853346 G allele and lower risk of gastric cancer. This association has been stronger in patients aged more than 60 years, non-smokers, non-drinkers, and those without family history of gastric cancer. Notably, expression assays have shown higher levels of PTENP1 in carriers of rs7853346 CG/GG genotype [37].
PTENP1 has also been shown to be down-regulated in osteoporosis patients, parallel with up-regulation of miR-214 [34].
Table 4
Dysregulation of PTENP1 in clinical samples
Tumor/ disease type
Numbers of clinical samples
Expression
(Tumor vs. normal)
Kaplan-Meier analysis
Polymorphism in PTENP1 associated with Tumor/ disease
Multivariate/ univariate cox regression
Clinicopathologic characteristics of patients
References
Acute leukemia
138 AL patients and 15 healthy controls
Downregulated
    
[10]
Bladder cancer
Plasma samples from 50 patients with bladder cancer and 60 normal subjects
20 pairs of tumor tissues and ANTs
Downregulated
   
High clinical grade
[11]
60 pairs of tumor tissues and ANTs
Upregulation of miR-20a (a target of PTENP1)
    
[12]
Breast cancer
52 pairs of tumor tissues and ANTs
Downregulated
Poorer OS
  
Advanced BC stages
[13]
65 pairs of tumor tissues and ANTs
Downregulated
    
[14]
20 pairs of tumor tissues and ANTs
Downregulated
    
[16]
Cervical cancer
54 pairs of tumor tissues and ANTs
Downregulated
   
FIGO stage and the lymph node metastasis
[17]
88 pairs of tumor tissues and ANTs
Downregulated
Poorer OS
  
Advanced stage, FIGO stage, tumor size and lymph node metastasis
[18]
56 pairs of tumor tissues and ANTs
Downregulated
Poorer OS
  
Advanced FIGO stage, metastasis and recurrence
[19]
Endometrioid endometrial carcinoma
40 pairs of tumor tissues and ANTs
GEO database and TGCA database
Downregulated
    
[20]
Esophageal carcinoma
GEO database (GSE20347): 17 pairs of tumor tissues and ANTs
Downregulated
    
[21]
93 ESCC patients
Downregulated
Poorer OS
 
TNM stage and PTENP1 expression were found to be independent factors that influence the OS of patients after radical esophagectomy.
Histological grade, more advanced TNM stage, deep infiltration depth, and lymph node metastasis
Gastric cancer
768 GC patients and 768 healthy controls
Downregulated
 
Patients who had rs7853346 G allele showed a remarkably decreased risk of GC in comparison with those carrying C allele.
Samples with rs7853346 CG/GG genotype showed high PTENP1 mRNA expression levels than those with CC genotype.
  
[37]
Gastric cancer
36 pairs of tumor tissues and ANTs
Downregulated
   
Tumor size, clinic stage and invasion depth
[22]
Glioma
23 gliomas tissue samples
Downregulated
    
[23]
279 glioma patients
Downregulated
 
Downregulated in patients carrying the CG&GG genotypes of rs7853346 compared with patients carrying the CC genotype of rs7853346
  
[38]
Head and neck squamous cell carcinoma
57 HNSCC tissues and 27 ANTs
Downregulated
Poorer OS or DFS
 
PTENP1 level was found to be an independent predictor of the OS and DFS in patients.
History of alcohol use
[25]
Hepatocellular carcinoma
48 pairs of tumor tissues and ANTs
Downregulated
Poorer OS
  
Tumor size and TNM stage
[27]
129 patients with HCC, 49 patients with liver cirrhosis, 27 patients with chronic HBV, and 93 normal subjects
Downregulated in HCC than in control groups
    
[39]
Multiple myeloma
43 multiple myeloma patients and 35 healthy controls
Upregulated
 
Samples with CC genotype showed higher levels of PTENP1 and TSC1 mRNA, and lower level of miR-19b compared to the CG and GG groups. G allele of rs7853346 polymorphism induces the proliferation of cancer stem cells.
  
[29]
Oral squamous cell carcinoma
62 pairs of tumor tissues and ANTs
Downregulated
Poorer OS
  
pT-stage and clinical stage
[30]
342 OSCC patients and 711 healthy controls
20 pairs of tumor tissues and ANTs
  
rs7853346 strongly reduced OSCC risk.
rs7853346 strongly decreased OSCC risk with gender, age, smoking and drinking condition adjusted.
 
[40]
Renal cell carcinoma
40 pairs of tumor tissues and ANTs
Downregulated
    
[31]
Osteoporosis
30 postmenopausal
females with osteoporosis and 15 premenopausal females with
arthritis (as controls)
Downregulated
    
[34]
ANTs adjacent non-cancerous tissues, OS overall survival, TNM tumor-node‐metastasis, HCC hepatocellular carcinoma, ESCC esophageal squamous cell carcinoma, HNSCC head and neck squamous cell carcinoma, DFS disease-free survival, AL acute leukemia

Discussion

PTENP1 is an lncRNA which primarily functions as a ceRNA to enhance expression of PTEN. This lncRNA acts as a sponge for some PETN-targeting miRNAs, such as miR-17, miR-20a, miR-19b, miR-106b, miR-200c, miR-193a-3p, miR-499-5p and miR-214. Besides, it can serve as a molecular sponge for other miRNAs such as miR-20a, miR-27a-3p, miR-17‐5p and miR-19b to influence expressions of PDCD4, EGR1, SOCS6 and TSC1, respectively.
The role of PTENP1 has been mostly evaluated in the pathoetiology of cancer. In this context, the results of in vitro, in vivo and clinical studies have been consistent. This lncRNA is regarded as a tumor suppressor lncRNA in all cancers except for multiple myeloma.
In addition, a number of investigations have shown its influence on development of non-malignant conditions such as alcohol-induced osteopenia, insulin resistance, osteoporosis, sepsis-associated cardiac dysfunction and spinal cord injury.
As an lncRNAs secreted in the exosomes, it has the potential to be used as a biomarker for early detection of cancers. This application has been evaluated in the context of bladder cancer. However, further studies in other cancers are needed to appraise the potential of PTENP1 in diagnostic purposes.
Although forced up-regulation of PTENP1 in cancer cell lines using different vectors could attenuate in vitro cancer cell proliferation and in vivo tumor growth, this field of study is still in its initial phases, needing further evaluations in animal models particularly focusing on bioavailability and biosafety issues. Additionally, a comprehensive evaluation of PTENP1 targets and related signaling pathways is necessary to avoid unwanted side effects.
Since up-regulation of PTENP1 can also enhance the cytotoxic effects of chemotherapeutic agents on cancer cells, therapies aimed at over-expression of this lncRNA are potential ways for combating chemoresistance.

Conclusions

Association between PTENP1 polymorphisms and susceptibility to cancer has been evaluated in Chinese gastric cancer patients. Additional studies in other types of cancers in different populations are needed to find the influence of genetic variants in this lncRNA on cancer risk.
Taken together, PTENP1 is an important modulator of cancer progression which not only increases expression of the important tumor suppressor PTEN, but also affects expression of other cancer-related genes such as those regulating cell cycle progression. Thus, this lncRNA represent a promising target for design of novel anti-cancer therapies.

Acknowledgements

This study was financially supported by Grant from Medical School of Shahid Beheshti University of Medical Sciences.

Declarations

All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards. Informed consent forms were obtained from all study participants. The study protocol was approved by the ethical committee of Shahid Beheshti University of Medical Sciences. All methods were performed in accordance with the relevant guidelines and regulations.
Not applicable.

Competing interests

The authors declare they have no conflict of interest.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Zhang X, Wang W, Zhu W, Dong J, Cheng Y, Yin Z, et al. Mechanisms and functions of long non-coding RNAs at multiple regulatory levels. Int J Mol Sci. 2019;20(22):5573.PubMedCentralCrossRef Zhang X, Wang W, Zhu W, Dong J, Cheng Y, Yin Z, et al. Mechanisms and functions of long non-coding RNAs at multiple regulatory levels. Int J Mol Sci. 2019;20(22):5573.PubMedCentralCrossRef
2.
Zurück zum Zitat Fatica A, Bozzoni I. Long non-coding RNAs: new players in cell differentiation and development. Nat Rev Genet. 2014;15(1):7–21.PubMedCrossRef Fatica A, Bozzoni I. Long non-coding RNAs: new players in cell differentiation and development. Nat Rev Genet. 2014;15(1):7–21.PubMedCrossRef
3.
4.
Zurück zum Zitat Yang G, Lu X, Yuan L. LncRNA: a link between RNA and cancer. Biochimica et Biophysica Acta (BBA). 2014;1839(11):1097–109.CrossRef Yang G, Lu X, Yuan L. LncRNA: a link between RNA and cancer. Biochimica et Biophysica Acta (BBA). 2014;1839(11):1097–109.CrossRef
5.
Zurück zum Zitat Statello L, Guo C-J, Chen L-L, Huarte M. Gene regulation by long non-coding RNAs and its biological functions. Nat Rev Mol Cell Biol. 2021;22(2):96–118.PubMedCrossRef Statello L, Guo C-J, Chen L-L, Huarte M. Gene regulation by long non-coding RNAs and its biological functions. Nat Rev Mol Cell Biol. 2021;22(2):96–118.PubMedCrossRef
6.
Zurück zum Zitat Ghafouri-Fard S, Khoshbakht T, Hussen BM, Taheri M, Mokhtari M. A review on the role of AFAP1-AS1 in the pathoetiology of cancer. Front Oncol. 2021;11:777849.PubMedPubMedCentralCrossRef Ghafouri-Fard S, Khoshbakht T, Hussen BM, Taheri M, Mokhtari M. A review on the role of AFAP1-AS1 in the pathoetiology of cancer. Front Oncol. 2021;11:777849.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Ghafouri-Fard S, Khoshbakht T, Taheri M, Jamali E. A concise review on the role of CircPVT1 in tumorigenesis, drug sensitivity, and cancer prognosis. Front Oncol. 2021;11:762960.PubMedPubMedCentralCrossRef Ghafouri-Fard S, Khoshbakht T, Taheri M, Jamali E. A concise review on the role of CircPVT1 in tumorigenesis, drug sensitivity, and cancer prognosis. Front Oncol. 2021;11:762960.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Ghafouri-Fard S, Khoshbakht T, Taheri M, Ebrahimzadeh K. A review on the carcinogenic roles of DSCAM-AS1. Front Cell Develop Biol. 2021;9:758513.CrossRef Ghafouri-Fard S, Khoshbakht T, Taheri M, Ebrahimzadeh K. A review on the carcinogenic roles of DSCAM-AS1. Front Cell Develop Biol. 2021;9:758513.CrossRef
9.
Zurück zum Zitat Wang Z. Antisense RNA and cancer. Cancer and noncoding RNAs. Amsterdam: Elsevier; 2018. p. 203–27.CrossRef Wang Z. Antisense RNA and cancer. Cancer and noncoding RNAs. Amsterdam: Elsevier; 2018. p. 203–27.CrossRef
10.
Zurück zum Zitat Wang C, Huai L, Zhang C, Jia Y, Li Q, Chen Y, et al. Study on expression of PTEN gene and its pseudogene PTENP1 in acute leukemia and correlation between them. Zhonghua xue ye xue za zhi Zhonghua xueyexue zazhi. 2012;33(11):896–901.PubMed Wang C, Huai L, Zhang C, Jia Y, Li Q, Chen Y, et al. Study on expression of PTEN gene and its pseudogene PTENP1 in acute leukemia and correlation between them. Zhonghua xue ye xue za zhi Zhonghua xueyexue zazhi. 2012;33(11):896–901.PubMed
11.
Zurück zum Zitat Zheng R, Du M, Wang X, Xu W, Liang J, Wang W, et al. Exosome–transmitted long non-coding RNA PTENP1 suppresses bladder cancer progression. Mol Cancer. 2018;17(1):1–13.CrossRef Zheng R, Du M, Wang X, Xu W, Liang J, Wang W, et al. Exosome–transmitted long non-coding RNA PTENP1 suppresses bladder cancer progression. Mol Cancer. 2018;17(1):1–13.CrossRef
12.
Zurück zum Zitat Zhong X, Wang L, Yan X, Yang X, Xiu H, Zhao M, et al. MiR-20a acted as a ceRNA of lncRNA PTENPL and promoted bladder cancer cell proliferation and migration by regulating PDCD4. Eur Rev Med Pharmacol Sci. 2020;24:2955–64.PubMed Zhong X, Wang L, Yan X, Yang X, Xiu H, Zhao M, et al. MiR-20a acted as a ceRNA of lncRNA PTENPL and promoted bladder cancer cell proliferation and migration by regulating PDCD4. Eur Rev Med Pharmacol Sci. 2020;24:2955–64.PubMed
13.
Zurück zum Zitat Gao X, Qin T, Mao J, Zhang J, Fan S, Lu Y, et al. PTENP1/miR-20a/PTEN axis contributes to breast cancer progression by regulating PTEN via PI3K/AKT pathway. J Exp Clin Cancer Res. 2019;38(1):1–14.CrossRef Gao X, Qin T, Mao J, Zhang J, Fan S, Lu Y, et al. PTENP1/miR-20a/PTEN axis contributes to breast cancer progression by regulating PTEN via PI3K/AKT pathway. J Exp Clin Cancer Res. 2019;38(1):1–14.CrossRef
14.
Zurück zum Zitat Li R, Guo L, Huang G, Luo W. PTENP1 acts as a ceRNA to regulate PTEN by sponging miR-19b and explores the biological role of PTENP1 in breast cancer. Cancer Gene Ther. 2017;24(7):309–15.PubMedCrossRef Li R, Guo L, Huang G, Luo W. PTENP1 acts as a ceRNA to regulate PTEN by sponging miR-19b and explores the biological role of PTENP1 in breast cancer. Cancer Gene Ther. 2017;24(7):309–15.PubMedCrossRef
15.
Zurück zum Zitat Chen S, Wang Y, Zhang J-H, Xia Q-J, Sun Q, Li Z-K, et al. Long non-coding RNA PTENP1 inhibits proliferation and migration of breast cancer cells via AKT and MAPK signaling pathways. Oncol Lett. 2017;14(4):4659–62.PubMedPubMedCentralCrossRef Chen S, Wang Y, Zhang J-H, Xia Q-J, Sun Q, Li Z-K, et al. Long non-coding RNA PTENP1 inhibits proliferation and migration of breast cancer cells via AKT and MAPK signaling pathways. Oncol Lett. 2017;14(4):4659–62.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Shi X, Tang X, Su L. Overexpression of long noncoding RNA PTENP1 inhibits cell proliferation and migration via suppression of miR-19b in breast cancer cells. Oncol Res. 2018;26(6):869.PubMedPubMedCentralCrossRef Shi X, Tang X, Su L. Overexpression of long noncoding RNA PTENP1 inhibits cell proliferation and migration via suppression of miR-19b in breast cancer cells. Oncol Res. 2018;26(6):869.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Fan Y, Sheng W, Meng Y, Cao Y, Li R. LncRNA PTENP1 inhibits cervical cancer progression by suppressing miR-106b. Artif cells Nanomed Biotechnol. 2020;48(1):393–407.PubMedCrossRef Fan Y, Sheng W, Meng Y, Cao Y, Li R. LncRNA PTENP1 inhibits cervical cancer progression by suppressing miR-106b. Artif cells Nanomed Biotechnol. 2020;48(1):393–407.PubMedCrossRef
19.
Zurück zum Zitat Ou L, Xiang T, Hao X, Wang D, Zeng Q. Reduced long non-coding RNA PTENP1 contributed to proliferation and invasion via miR-19b/MTUS1 axis in patients with cervical cancer. Eur Rev Med Pharmacol Sci. 2020;24:4132–44.PubMed Ou L, Xiang T, Hao X, Wang D, Zeng Q. Reduced long non-coding RNA PTENP1 contributed to proliferation and invasion via miR-19b/MTUS1 axis in patients with cervical cancer. Eur Rev Med Pharmacol Sci. 2020;24:4132–44.PubMed
20.
Zurück zum Zitat Chen R, Zhang M, Liu W, Chen H, Cai T, Xiong H, et al. Estrogen affects the negative feedback loop of PTENP1-miR200c to inhibit PTEN expression in the development of endometrioid endometrial carcinoma. Cell Death Dis. 2018;10(1):1–13. Chen R, Zhang M, Liu W, Chen H, Cai T, Xiong H, et al. Estrogen affects the negative feedback loop of PTENP1-miR200c to inhibit PTEN expression in the development of endometrioid endometrial carcinoma. Cell Death Dis. 2018;10(1):1–13.
21.
Zurück zum Zitat Gong T, Zheng S, Huang S, Fu S, Zhang X, Pan S, et al. PTENP1 inhibits the growth of esophageal squamous cell carcinoma by regulating SOCS6 expression and correlates with disease prognosis. Mol Carcinog. 2017;56(12):2610–9.PubMedPubMedCentralCrossRef Gong T, Zheng S, Huang S, Fu S, Zhang X, Pan S, et al. PTENP1 inhibits the growth of esophageal squamous cell carcinoma by regulating SOCS6 expression and correlates with disease prognosis. Mol Carcinog. 2017;56(12):2610–9.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Zhang R, Guo Y, Ma Z, Ma G, Xue Q, Li F, et al. Long non-coding RNA PTENP1 functions as a ceRNA to modulate PTEN level by decoying miR-106b and miR-93 in gastric cancer. Oncotarget. 2017;8(16):26079.PubMedPubMedCentralCrossRef Zhang R, Guo Y, Ma Z, Ma G, Xue Q, Li F, et al. Long non-coding RNA PTENP1 functions as a ceRNA to modulate PTEN level by decoying miR-106b and miR-93 in gastric cancer. Oncotarget. 2017;8(16):26079.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Hu S, Xu L, Li L, Luo D, Zhao H, Li D, et al. Overexpression of lncRNA PTENP1 suppresses glioma cell proliferation and metastasis in vitro. Onco Targets Ther. 2019;12:147.PubMedCrossRef Hu S, Xu L, Li L, Luo D, Zhao H, Li D, et al. Overexpression of lncRNA PTENP1 suppresses glioma cell proliferation and metastasis in vitro. Onco Targets  Ther. 2019;12:147.PubMedCrossRef
24.
Zurück zum Zitat Hao S, Ma H, Niu Z, Sun S, Zou Y, Xia H. hUC-MSCs secreted exosomes inhibit the glioma cell progression through PTENP1/miR-10a-5p/PTEN pathway. Eur Rev Med Pharmacol Sci. 2019;23(22):10013–23.PubMed Hao S, Ma H, Niu Z, Sun S, Zou Y, Xia H. hUC-MSCs secreted exosomes inhibit the glioma cell progression through PTENP1/miR-10a-5p/PTEN pathway. Eur Rev Med Pharmacol Sci. 2019;23(22):10013–23.PubMed
25.
Zurück zum Zitat Liu J, Xing Y, Xu L, Chen W, Cao W, Zhang C. Decreased expression of pseudogene PTENP1 promotes malignant behaviours and is associated with the poor survival of patients with HNSCC. Sci Rep. 2017;7(1):1–11.CrossRef Liu J, Xing Y, Xu L, Chen W, Cao W, Zhang C. Decreased expression of pseudogene PTENP1 promotes malignant behaviours and is associated with the poor survival of patients with HNSCC. Sci Rep. 2017;7(1):1–11.CrossRef
26.
Zurück zum Zitat Cao L-q, Yang X-w, Chen Y-b, Zhang D-w, Jiang X-F, Xue P. Exosomal miR-21 regulates the TETs/PTENp1/PTEN pathway to promote hepatocellular carcinoma growth. Mol Cancer. 2019;18(1):1–14.CrossRef Cao L-q, Yang X-w, Chen Y-b, Zhang D-w, Jiang X-F, Xue P. Exosomal miR-21 regulates the TETs/PTENp1/PTEN pathway to promote hepatocellular carcinoma growth. Mol Cancer. 2019;18(1):1–14.CrossRef
27.
Zurück zum Zitat Qian Y-Y, Li K, Liu Q-Y, Liu Z-S. Long non-coding RNA PTENP1 interacts with miR-193a-3p to suppress cell migration and invasion through the PTEN pathway in hepatocellular carcinoma. Oncotarget. 2017;8(64):107859.PubMedPubMedCentralCrossRef Qian Y-Y, Li K, Liu Q-Y, Liu Z-S. Long non-coding RNA PTENP1 interacts with miR-193a-3p to suppress cell migration and invasion through the PTEN pathway in hepatocellular carcinoma. Oncotarget. 2017;8(64):107859.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Chen C-L, Tseng Y-W, Wu J-C, Chen G-Y, Lin K-C, Hwang S-M, et al. Suppression of hepatocellular carcinoma by baculovirus-mediated expression of long non-coding RNA PTENP1 and MicroRNA regulation. Biomaterials. 2015;44:71–81.PubMedCrossRef Chen C-L, Tseng Y-W, Wu J-C, Chen G-Y, Lin K-C, Hwang S-M, et al. Suppression of hepatocellular carcinoma by baculovirus-mediated expression of long non-coding RNA PTENP1 and MicroRNA regulation. Biomaterials. 2015;44:71–81.PubMedCrossRef
29.
Zurück zum Zitat Zhang Y, Xu C. G allele of rs7853346 polymorphism in PTENP1 enhances the proliferation of multiple myeloma cancer stem cells by promoting the expression of PTENP1 and its downstream signaling molecules. J Cell Biochem. 2019;120(12):19738–48.PubMedCrossRef Zhang Y, Xu C. G allele of rs7853346 polymorphism in PTENP1 enhances the proliferation of multiple myeloma cancer stem cells by promoting the expression of PTENP1 and its downstream signaling molecules. J Cell Biochem. 2019;120(12):19738–48.PubMedCrossRef
30.
Zurück zum Zitat Gao L, Ren W, Zhang L, Li S, Kong X, Zhang H, et al. PTENp1, a natural sponge of miR-21, mediates PTEN expression to inhibit the proliferation of oral squamous cell carcinoma. Mol Carcinog. 2017;56(4):1322–34.PubMedCrossRef Gao L, Ren W, Zhang L, Li S, Kong X, Zhang H, et al. PTENp1, a natural sponge of miR-21, mediates PTEN expression to inhibit the proliferation of oral squamous cell carcinoma. Mol Carcinog. 2017;56(4):1322–34.PubMedCrossRef
31.
Zurück zum Zitat Yu G, Yao W, Gumireddy K, Li A, Wang J, Xiao W, et al. Pseudogene PTENP1 functions as a competing endogenous RNA to suppress clear-cell renal cell carcinoma progression. Mol Cancer Ther. 2014;13(12):3086–97.PubMedPubMedCentralCrossRef Yu G, Yao W, Gumireddy K, Li A, Wang J, Xiao W, et al. Pseudogene PTENP1 functions as a competing endogenous RNA to suppress clear-cell renal cell carcinoma progression. Mol Cancer Ther. 2014;13(12):3086–97.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Chen YX, Zhu DY, Gao J, Xu ZL, Tao SC, Yin WJ, et al. Diminished membrane recruitment of Akt is instrumental in alcohol-associated osteopenia via the PTEN/Akt/GSK‐3β/β‐catenin axis. FEBS J. 2019;286(6):1101–19.PubMedCrossRef Chen YX, Zhu DY, Gao J, Xu ZL, Tao SC, Yin WJ, et al. Diminished membrane recruitment of Akt is instrumental in alcohol-associated osteopenia via the PTEN/Akt/GSK‐3β/β‐catenin axis. FEBS J. 2019;286(6):1101–19.PubMedCrossRef
33.
Zurück zum Zitat Wang L, Zhang N, Wang Z, Ai D-m, Cao Z-y, Pan H-p. Pseudogene PTENP1 functions as a competing endogenous RNA (ceRNA) to regulate PTEN expression by sponging miR-499-5p. Biochemistry (Moscow). 2016;81(7):739–47.CrossRef Wang L, Zhang N, Wang Z, Ai D-m, Cao Z-y, Pan H-p. Pseudogene PTENP1 functions as a competing endogenous RNA (ceRNA) to regulate PTEN expression by sponging miR-499-5p. Biochemistry (Moscow). 2016;81(7):739–47.CrossRef
34.
Zurück zum Zitat Wang C-G, Wang L, Yang T, Su S-L, Hu Y-H, Zhong D. Pseudogene PTENP1 sponges miR-214 to regulate the expression of PTEN to modulate osteoclast differentiation and attenuate osteoporosis. Cytotherapy. 2020;22(8):412–23.PubMedCrossRef Wang C-G, Wang L, Yang T, Su S-L, Hu Y-H, Zhong D. Pseudogene PTENP1 sponges miR-214 to regulate the expression of PTEN to modulate osteoclast differentiation and attenuate osteoporosis. Cytotherapy. 2020;22(8):412–23.PubMedCrossRef
35.
Zurück zum Zitat Liu Y, Liu L, Zhang J. Protective role of matrine in sepsis-associated cardiac dysfunction through regulating the lncRNA PTENP1/miR-106b-5p axis. Biomed Pharmacother. 2021;134:111112.PubMedCrossRef Liu Y, Liu L, Zhang J. Protective role of matrine in sepsis-associated cardiac dysfunction through regulating the lncRNA PTENP1/miR-106b-5p axis. Biomed Pharmacother. 2021;134:111112.PubMedCrossRef
36.
Zurück zum Zitat Yuan M, Zhao S, Chen R, Wang G, Bie Y, Wu Q, et al. MicroRNA–138 inhibits tumor growth and enhances chemosensitivity in human cervical cancer by targeting H2AX. Exp Ther Med. 2020;19(1):630–8.PubMed Yuan M, Zhao S, Chen R, Wang G, Bie Y, Wu Q, et al. MicroRNA–138 inhibits tumor growth and enhances chemosensitivity in human cervical cancer by targeting H2AX. Exp Ther Med. 2020;19(1):630–8.PubMed
37.
Zurück zum Zitat Ge Y, He Y, Jiang M, Luo D, Huan X, Wang W, et al. Polymorphisms in lncRNA PTENP1 and the risk of gastric cancer in a Chinese population. Dis Markers. 2017;2017:6807452.PubMedPubMedCentralCrossRef Ge Y, He Y, Jiang M, Luo D, Huan X, Wang W, et al. Polymorphisms in lncRNA PTENP1 and the risk of gastric cancer in a Chinese population. Dis Markers. 2017;2017:6807452.PubMedPubMedCentralCrossRef
38.
39.
Zurück zum Zitat Huang J, Zheng Y, Xiao X, Liu C, Lin J, Zheng S, et al. A circulating long noncoding RNA panel serves as a diagnostic marker for hepatocellular carcinoma. Dis Markers. 2020;2020:5417598.PubMedPubMedCentral Huang J, Zheng Y, Xiao X, Liu C, Lin J, Zheng S, et al. A circulating long noncoding RNA panel serves as a diagnostic marker for hepatocellular carcinoma. Dis Markers. 2020;2020:5417598.PubMedPubMedCentral
40.
Zurück zum Zitat Xin C, Li J, Zhang Y, Yu Z. Polymorphisms in lncRNA PTENP1 and the risk of oral squamous cell carcinoma in a Chinese population. Eur Rev Med Pharmacol Sci. 2018;22(17):5583–7.PubMed Xin C, Li J, Zhang Y, Yu Z. Polymorphisms in lncRNA PTENP1 and the risk of oral squamous cell carcinoma in a Chinese population. Eur Rev Med Pharmacol Sci. 2018;22(17):5583–7.PubMed
Metadaten
Titel
A review on the role of PTENP1 in human disorders with an especial focus on tumor suppressor role of this lncRNA
verfasst von
Soudeh Ghafouri-Fard
Tayyebeh Khoshbakht
Bashdar Mahmud Hussen
Mohammad Taheri
Nader Akbari Dilmaghani
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Erschienen in
Cancer Cell International / Ausgabe 1/2022
Elektronische ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-022-02625-8

Weitere Artikel der Ausgabe 1/2022

Cancer Cell International 1/2022 Zur Ausgabe

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Labor, CT-Anthropometrie zeigen Risiko für Pankreaskrebs

13.05.2024 Pankreaskarzinom Nachrichten

Gerade bei aggressiven Malignomen wie dem duktalen Adenokarzinom des Pankreas könnte Früherkennung die Therapiechancen verbessern. Noch jedoch klafft hier eine Lücke. Ein Studienteam hat einen Weg gesucht, sie zu schließen.

Viel pflanzliche Nahrung, seltener Prostata-Ca.-Progression

12.05.2024 Prostatakarzinom Nachrichten

Ein hoher Anteil pflanzlicher Nahrung trägt möglicherweise dazu bei, das Progressionsrisiko von Männern mit Prostatakarzinomen zu senken. In einer US-Studie war das Risiko bei ausgeprägter pflanzlicher Ernährung in etwa halbiert.

Alter verschlechtert Prognose bei Endometriumkarzinom

11.05.2024 Endometriumkarzinom Nachrichten

Ein höheres Alter bei der Diagnose eines Endometriumkarzinoms ist mit aggressiveren Tumorcharakteristika assoziiert, scheint aber auch unabhängig von bekannten Risikofaktoren die Prognose der Erkrankung zu verschlimmern.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.