Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
1.
Biochem Pharmacol ; 222: 116109, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38458330

RESUMEN

Angiotensin (Ang)-(1-7) stimulates vasoprotective functions of diabetic (DB) CD34+ hematopoietic stem/progenitor cells partly by decreasing reactive oxygen species (ROS), increasing nitric oxide (NO) levels and decreasing TGFß1 secretion. Telomerase reverse transcriptase (TERT) translocates to mitochondria and regulates ROS generation. Alternative splicing of TERT results in variants α-, ß- and α-ß-TERT, which may oppose functions of full-length (FL) TERT. This study tested if the protective functions of Ang-(1-7) or TGFß1-silencing are mediated by mitoTERT and that diabetes decreases FL-TERT expression by inducing splicing. CD34+ cells were isolated from the peripheral blood mononuclear cells of nondiabetic (ND, n = 68) or DB (n = 74) subjects. NO and mitoROS levels were evaluated by flow cytometry. TERT splice variants and mitoDNA-lesions were characterized by qPCR. TRAP assay was used for telomerase activity. Decoy peptide was used to block mitochondrial translocation (mitoXTERT). TERT inhibitor or mitoXTERT prevented the effects of Ang-(1-7) on NO or mitoROS levels in DB-CD34+ cells. FL-TERT expression and telomerase activity were lower and mitoDNA-lesions were higher in DB cells compared to ND and were reversed by Ang-(1-7) or TGFß1-silencing. The prevalence of TERT splice variants, with predominant ß-TERT expression, was higher and the expression of FL-TERT was lower in DB cells (n = 25) compared to ND (n = 30). Ang-(1-7) or TGFß1-silencing decreased TERT-splicing and increased FL-TERT. Blocking of ß-splicing increased FL-TERT and protected mitoDNA in DB-cells. The findings suggest that diabetes induces TERT-splicing in CD34+ cells and that ß-TERT splice variant largely contributes to the mitoDNA oxidative damage.


Asunto(s)
Angiotensina I , Diabetes Mellitus , Fragmentos de Péptidos , Telomerasa , Humanos , Telomerasa/genética , Telomerasa/metabolismo , Telomerasa/farmacología , Especies Reactivas de Oxígeno/metabolismo , Leucocitos Mononucleares , Mitocondrias/metabolismo , Diabetes Mellitus/metabolismo
2.
FEBS Open Bio ; 13(9): 1667-1682, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37525387

RESUMEN

Telomerase reverse transcriptase (TERT) is a protein that catalyzes the reverse transcription of telomere elongation. TERT is also expected to play a non-canonical role beyond telomere lengthening since it localizes not only in the nucleus but also in mitochondria, where telomeres do not exist. Several studies have reported that mitochondrial TERT regulates apoptosis induced by oxidative stress. However, there is still some controversy as to whether mitochondrial TERT promotes or inhibits apoptosis, mainly due to the lack of information on changes in TERT distribution in individual cells over time. Here, we simultaneously detected apoptosis and TERT localization after oxidative stress in individual HeLa cells by live-cell tracking. Single-cell tracking revealed that the stress-induced accumulation of TERT in mitochondria caused apoptosis, but that accumulation increased over time until cell death. The results suggest a new model in which mitochondrial TERT has two opposing effects at different stages of apoptosis: it predetermines apoptosis at the first stage of cell-fate determination, but also delays apoptosis at the second stage. As such, our data support a model that integrates the two opposing hypotheses on mitochondrial TERT's effect on apoptosis. Furthermore, detailed statistical analysis of TERT mutations, which have been predicted to inhibit TERT transport to mitochondria, revealed that these mutations suppress apoptosis independent of mitochondrial localization of TERT. Together, these results imply that the non-canonical functions of TERT affect a wide range of mitochondria-dependent and mitochondria-independent apoptosis pathways.


Asunto(s)
Mitocondrias , Telomerasa , Humanos , Células HeLa , Mitocondrias/metabolismo , Telomerasa/genética , Telomerasa/metabolismo , Telomerasa/farmacología , Núcleo Celular/metabolismo , ARN Polimerasas Dirigidas por ADN , Apoptosis
3.
Gynecol Obstet Invest ; 88(4): 226-239, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37429261

RESUMEN

OBJECTIVES: The effect of telomerase inhibitor BIBR1532 on endometriotic cells was investigated to explore the inhibitory effect of targeting telomerase on endometriosis. DESIGN: In vitro primary cell culture study. Participants/Materials: Primary endometrial cells derived from eutopic and ectopic endometrium in patients with endometriosis. SETTING: The study was conducted in the university hospital. METHODS: Paired eutopic and ectopic endometrial cells were collected from 6 patients from January 2018 to July 2021. A TRAP assay was performed to detect the telomerase activity of the cells. MTT, cell cycle, apoptosis, migration, and invasion assays were performed to study the inhibitory effect of BIBR1532. Enrichment analysis was performed to identify the key pathways involved in endometriosis progression and telomerase action. Then, Western blotting was used to investigate the expression of related proteins. RESULTS: BIBR1532 treatment significantly inhibited the growth of eutopic and ectopic endometrial cells, with apoptosis and cell cycle signaling involved. Migration and invasion, important characteristics for the establishment of ectopic lesions, were also inhibited by BIBR1532. The MAPK signaling cascade, related to telomerase and endometriosis, was decreased in eutopic and ectopic endometrial stromal cells with the treatment of BIBR1532. LIMITATIONS: The severe side effects of telomerase inhibitors might be the main obstacle to clinical application, so it is necessary to find better drug delivery methods in vivo. CONCLUSIONS: The telomerase inhibitor BIBR1532 affects endometrial cell proliferation, migration, and invasion in endometriosis.


Asunto(s)
Hiperplasia Endometrial , Endometriosis , Telomerasa , Femenino , Humanos , Endometriosis/patología , Telomerasa/metabolismo , Telomerasa/farmacología , Aminobenzoatos/metabolismo , Aminobenzoatos/farmacología , Hiperplasia Endometrial/patología , Endometrio/patología , Proliferación Celular , Células del Estroma/metabolismo
4.
Reprod Fertil Dev ; 35(10): 527-538, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37263290

RESUMEN

CONTEXT: Telomerase reverse transcriptase is a key factor responsible for structural and cellular alterations in aged oocytes and changes in the structure of the zona pellucida and mitochondria. Telomerase expression is reduced in aged cumulus oocyte complexes, and its activation or enhanced expression would be beneficial for in vitro oocyte maturation and in vitro embryo development. AIMS: This study aimed to investigate telomerase activation by cycloastragenol and its effect on bovine oocyte in vitro maturation, fertilisation, and early embryo development. METHODS: We used qPCR, Western blot, immunofluorescence, reactive oxygen species (ROS) assay,TUNEL assay, JC-1 assay, and invasion assay to analyse the affect of cycloastragenol (CAG) on bovine oocyte maturation, embryo development, embryo quality and implantation potential. KEY RESULTS: Cycloastragenol treatment of oocytes in in vitro maturation (IVM) media significantly (P <0.05) improved oocyte IVM (90.87%), embryo cleavage (90.78%), blastocyst hatching (27.04%), and embryo implantation potential. Telomerase also interacts with mitochondria, and JC-1 staining results showed significantly (P <0.05) higher mitochondrial membrane potential (ΔΨ m) in the CAG-treated group. Furthermore, the inner cell mass (OCT4 and SOX2) and trophoblasts (CDX2) of the control and CAG groups were examined. Moreover, CAG treatment to primary cultured bovine cumulus cells substantially enhanced telomerase activity. CONCLUSIONS: Telomerase activation via cycloastragenol is beneficial for bovine oocyte IVM and for the production of high-quality bovine embryos. IMPLICATIONS: Cycloastragenol is a natural telomerase activator, and could be useful as a permanent component of oocyte maturation media.


Asunto(s)
Telomerasa , Femenino , Animales , Bovinos , Telomerasa/genética , Telomerasa/metabolismo , Telomerasa/farmacología , Células del Cúmulo/metabolismo , Oocitos/metabolismo , Técnicas de Maduración In Vitro de los Oocitos/veterinaria , Técnicas de Maduración In Vitro de los Oocitos/métodos , Implantación del Embrión , Desarrollo Embrionario , Blastocisto
5.
Chin J Physiol ; 66(3): 119-128, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37322622

RESUMEN

Glioblastoma (GB) is one of the most aggressive and malignant tumors of the central nervous system. Conventional treatment for GB requires surgical resection followed by radiotherapy combined with temozolomide chemotherapy; however, the median survival time is only 12-15 months. Angelica sinensis Radix (AS) is commonly used as a traditional medicinal herb or a food/dietary supplement in Asia, Europe, and North America. This study aimed to investigate the effect of AS-acetone extract (AS-A) on the progression of GB and the potential mechanisms underlying its effects. The results indicated that AS-A used in this study showed potency in growth inhibition of GB cells and reduction of telomerase activity. In addition, AS-A blocked the cell cycle at the G0/G1 phase by regulating the expression of p53 and p16. Furthermore, apoptotic morphology, such as chromatin condensation, DNA fragmentation, and apoptotic bodies, was observed in AS-A-treated cells, induced by the activation of the mitochondria-mediated pathway. In an animal study, AS-A reduced tumor volume and prolonged lifespans of mice, with no significant changes in body weight or obvious organ toxicity. This study confirmed the anticancer effects of AS-A by inhibiting cell proliferation, reducing telomerase activity, altering cell cycle progression, and inducing apoptosis. These findings suggest that AS-A has great potential for development as a novel agent or dietary supplement against GB.


Asunto(s)
Glioblastoma , Telomerasa , Humanos , Ratones , Animales , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Glioblastoma/patología , Telomerasa/metabolismo , Telomerasa/farmacología , Telomerasa/uso terapéutico , Apoptosis , Puntos de Control del Ciclo Celular , Ciclo Celular , Proliferación Celular , Telómero/metabolismo , Telómero/patología , Mitocondrias , Línea Celular Tumoral
6.
Eur Spine J ; 32(7): 2431-2440, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37165116

RESUMEN

PURPOSE: To investigate the interaction of telomerase activity and telomere length on neuro-protection or neuro-degeneration effects after spinal cord injury (SCI). METHODS: A contusive SCI model was developed using 56 Sprague-Dawley rats. Seven rats were allocated into acute injury phase groups (1, 3, 8, 24, and 48 h), and sub-acute and chronic injury phase groups (1, 2, and 4 weeks). Telomerase activity was assessed by telomerase reverse transcriptase (TERT) and telomeric repeat binding factor-2 (TERF-2). Differentiation of activated neural stem cells was investigated by co-expression of neuronal/glial cell markers. Apoptosis expression was also investigated by caspase-3, 8, and 9 using terminal deoxynucleotidyl transferase dUTP nick end labelling staining. Immunofluorescence staining and western blotting were performed for quantitative analyses. RESULTS: Expression of TERT increased gradually until 24 h post-injury, and was decreased following SCI (P < 0.05). TERF-2 also was increased following SCI until 24 h post-injury and then decreased with time (P < 0.05). Co-localization of TERT and TERF-2 was higher at 24 h post-injury. High expression of TERT was seen in neurons (Neu N Ab), however, expression of TERT was relatively lower in astrocytes and oligodendrocytes. Apoptosis analysis showed persistent high expression of caspases-3, -9, and -8 during the observation period. CONCLUSIONS: Increased TERT and TERF-2 activity were noted 24 h post-injury in the acute phase of SCI with TERF-2 maintaining telomeric-repeat length. Our results suggest that increased activity of telomere maintenance may be related to neuro-protective mechanisms against subsequent apoptosis resulting from DNA damage after acute SCI.


Asunto(s)
Traumatismos de la Médula Espinal , Telomerasa , Ratas , Animales , Ratas Sprague-Dawley , Telomerasa/genética , Telomerasa/metabolismo , Telomerasa/farmacología , Apoptosis , Neuronas/metabolismo , Médula Espinal/metabolismo
7.
Comb Chem High Throughput Screen ; 26(14): 2476-2486, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37073660

RESUMEN

AIM AND OBJECTIVE: Long intergenic non-coding RNA-p21 (lincRNA-p21) plays a critical role in various senescence-associated physiological and pathological conditions. We aimed to explore the senescence-associated effects of lincRNA-p21 in 1-methyl-4-phenylpyridinium (MPP+) treated neuroblastoma SH-SY5Y cell line as a therapeutic target. MATERIALS AND METHODS: The RNA expression levels of lincRNA-p21, p53, p16, and telomere length were examined with reverse transcription-quantitative polymerase chain reaction (RTqPCR). The Telo TAGGG™ Telomerase PCR ELISA PLUS Kit was used to determine telomerase activity. Cellular viability was evaluated with the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and lactate dehydrogenase (LDH) assay. Western blot was performed to analyze ß-catenin protein expression. Besides, oxidative stress was evaluated by Jaggregate- forming delocalized lipophilic cation, 5,5',6,6'-tetrachloro-1,1',3,3'- tetraethylbenzimidazolocarbocyanine++ + iodide (JC­1) stain, fluorescence spectrophotometry, colorimetric assay, and malondialdehyde (MDA) formation. RESULTS: This research demonstrated that MPP+ caused a distinct increase in the expression of LincRNA- p21 in SH-SY5Y cells. MPP+ induced cellular senescence with decreasing cellular proliferation and viability, increasing expression levels of senescence-associated makers such as genes p53 and p16, accompanied by significantly decreasing telomere length and telomerase activity. At the same time, these effects were abolished by silencing lincRNA-p21 with small interfering RNA (siRNA). On the contrary, ß-catenin silencing contributes to reversing anti-senescent effects caused by lincRNA-p21 silencing. Moreover, modifying lincRNA-p21 exerted an anti-senescent influence depending on decreasing oxidant stress. CONCLUSION: Our study showed that in the treatment of MPP+, lincRNA-p21 might serve a role in the SH-SY5Y cell senescence by modulating the Wnt/ß-catenin pathway, as well as increasing oxidant stress. Thus, trying to target lincRNA-p21 may have important therapeutic and practical implications for PD.


Asunto(s)
Neuroblastoma , ARN Largo no Codificante , Telomerasa , Humanos , 1-Metil-4-fenilpiridinio/farmacología , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Telomerasa/metabolismo , Telomerasa/farmacología , beta Catenina/metabolismo , Proteína p53 Supresora de Tumor/genética , Apoptosis , Senescencia Celular , Oxidantes/farmacología , Línea Celular Tumoral
8.
Arch Immunol Ther Exp (Warsz) ; 71(1): 6, 2023 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-36807774

RESUMEN

Colon cancer etiology involves a wide spectrum of genetic and epigenetic alterations, finding it challenging to find effective therapeutic strategies. Quercetin exhibits potent anti-proliferative/apoptotic properties. In the present study, we aimed to elucidate the anti-cancer and anti-aging effect of quercetin in colon cancer cell lines. The anti-proliferative effect of quercetin was assessed in vitro by CCK-8 in normal and colon cancer cell lines. To check the anti-aging potential of quercetin, collagenase, elastase, and hyaluronidase inhibitory activity assays were performed. The epigenetic and DNA damage assays were performed using the human NAD-dependent deacetylase Sirtuin-6, proteasome 20S, Klotho, Cytochrome-C, and telomerase ELISA kits. Furthermore, the aging-associated miRNA expression profiling was performed on colon cancer cells. The treatment with quercetin inhibited cell proliferation of colon cancer cells in a dose-dependent manner. Quercetin arrested colon cancer cell growth by modulating expression of aging proteins including Sirtuin-6 and Klotho and also by inhibiting telomerase activity to restrict the telomere length which is evident from qPCR analysis. Quercetin also exhibited DNA damage protection by reducing proteasome 20S levels. The miRNA expression profiling results displayed differential expression of miRNA in colon cancer cell, and in addition, the highly upregulated miRNA was involved in the regulation of cell cycle, proliferation, and transcription. Our data suggest that quercetin treatment inhibited cell proliferation in colon cancer cells through regulating the anti-aging protein expression and provides better understanding for quercetin's potential use in colon cancer treatment.


Asunto(s)
Neoplasias del Colon , MicroARNs , Sirtuinas , Telomerasa , Humanos , Apoptosis , Proliferación Celular , Epigénesis Genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Complejo de la Endopetidasa Proteasomal/farmacología , Complejo de la Endopetidasa Proteasomal/uso terapéutico , Quercetina/farmacología , Quercetina/uso terapéutico , Sirtuinas/metabolismo , Sirtuinas/farmacología , Sirtuinas/uso terapéutico , Telomerasa/metabolismo , Telomerasa/farmacología , Telomerasa/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico
9.
Metab Brain Dis ; 38(3): 1067-1077, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36287355

RESUMEN

Aging is widely thought to be associated with oxidative stress. Momordica charantia (MC) is a classic vegetable and traditional herbal medicine widely consumed in Asia, and M. charantia polysaccharide (MCP) is the main bioactive ingredient of MC. We previously reported an antioxidative and neuroprotective effect of MCP in models of cerebral ischemia/reperfusion and hemorrhage injury. However, the role played by MCP in neurodegenerative diseases, especially during aging, remains unknown. In this study, we investigated the protective effect of MCP against oxidative stress and brain damage in a D-galactose-induced aging model (DGAM). The Morris water maze test was performed to evaluate the spatial memory function of model rats. The levels of malondialdehyde (MDA), glutathione (GSH), and superoxide dismutase (SOD) were measured and telomerase activity was determined. The results showed that MCP treatment attenuated spatial memory dysfunction induced by D-galactose. In addition, MCP increased antioxidant capacity by decreasing MDA and increasing SOD and GSH levels. MCP treatment also improved telomerase activity in aging rats. Mechanistically, MCP promoted the entry of both Nrf2 and ß-Catenin into the nucleus, which is the hallmark of antioxidation signaling pathway activation. This study highlights a role played by MCP in ameliorating aging-induced oxidative stress injury and reversing the decline in learning and memory capacity. Our work provides evidence that MCP administration might be a potential antiaging strategy.


Asunto(s)
Momordica charantia , Telomerasa , Ratas , Animales , Galactosa/toxicidad , Momordica charantia/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , beta Catenina/metabolismo , Telomerasa/metabolismo , Telomerasa/farmacología , Envejecimiento/metabolismo , Estrés Oxidativo , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Antioxidantes/metabolismo , Transducción de Señal , Polisacáridos/farmacología , Polisacáridos/uso terapéutico , Superóxido Dismutasa/metabolismo , Malondialdehído/metabolismo
10.
Arch Biochem Biophys ; 722: 109218, 2022 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-35430213

RESUMEN

BACKGROUND: Angiotensin (Ang) (1-7) is a vasodilator peptide that ameliorates microcirculation dysfunction, increases telomerase activity in cells, and exerts vasodilatory, anti-inflammatory, antioxidative stress, and antiapoptotic effects. Mitochondrial human telomerase reverse transcriptase (hTERT) plays an important role in the processes of antiapoptosis, antioxidative stress, and immortalization. This study aimed to investigate the effect of Ang(1-7) on the mitochondrial translocation of hTERT. METHODS: An in vitro model of lipopolysaccharide (LPS)-induced inflammation was established in human umbilical vein endothelial cells (HUVECs). Ang(1-7) was added to cells 30 min before LPS stimulation. The Ang(1-7)/Mas receptor antagonist A779 plus Ang(1-7) were added to the cells 30 min before LPS stimulation. The translocase outer membrane (TOM)20-overexpression HUVECs (HUVEC-TOM20OE), TOM20-knockdown HUVECs (HUVEC-TOM20KD), and the corresponding negative control cell lines were constructed by lentiviral transfection of HUVECs. Cells subjected to LPS stimulation alone, LPS plus Ang(1-7), LPS plus Ang(1-7) and A779, vehicle and no treatment were termed the LPS group, LPS + A group, LPS + A + A779 group, Con group and Neg group, respectively. Immunofluorescence staining was used to detect the distribution of hTERT in the nuclei and mitochondria of HUVECs and to locate TOM20, TOM40, and translocase inner membrane (TIM)23 in the mitochondria. The protein expression levels of total hTERT, mitochondrial hTERT, TOM20, TOM40, and TIM23 were measured by Western blot. The mRNA expression levels of hTERT, TOM20, TOM40, and TIM23 were assessed by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). RESULTS: hTERT colocalized with TOM40, TOM20 and TIM23 in the mitochondria. The mitochondrial hTERT protein level of the LPS + A group was significantly greater than that of the LPS group (P = 0.001), and the LPS group showed significantly increased expression of mitochondrial hTERT compared with that of the control group (P = 0.001). No significant difference in the level of total hTERT was observed between the LPS + A and LPS groups. The mitochondrial hTERT protein level of the LPS + A + A779 group was significantly lower than that of the LPS + A group (P = 0.021). The protein level of mitochondrial hTERT in HUVEC-TOM20KD treated with or without LPS alone or LPS + A was significantly decreased compared with the corresponding groups of control HUVECs (HUVEC-TOM20KD-Con vs. HUVEC-Con, P = 0.035; HUVEC-TOM20KD-LPS vs. HUVEC-LPS, P = 0.003; HUVEC-TOM20KD-LPS + A vs. HUVEC-LPS + A, P = 0.001), and treatment with Ang(1-7) did not restore the downregulation of mitochondrial hTERT in HUVEC-TOM20KD. HUVEC-TOM20OE showed a significantly increased level of mitochondrial hTERT (HUVEC-TOM20OE-Con vs. HUVEC-Con, P = 0.010), which was further elevated by Ang(1-7) stimulation (HUVEC-TOM20OE-LPS + A vs. HUVEC-TOM20OE-Con, P = 0.011). Lastly, the protein expression levels of TOM40 (HUVEC-TOM20KD-Con vs. HUVEC-Con, P = 0.007) and TIM23 (HUVEC-TOM20KD-Con vs. HUVEC-Con, P = 0.001) were significantly increased in HUVEC-TOM20KD in comparison to HUVECs. CONCLUSIONS: Ang(1-7) effectively promoted mitochondrial translocation of hTERT in HUVECs via TOM20, indicating that hTERT may be transported to the mitochondria through the TOM20 complex. In addition, A779 could block the effects of Ang(1-7) in HUVECs.


Asunto(s)
Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales/metabolismo , Telomerasa , Angiotensina I , Angiotensina II/farmacología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Lipopolisacáridos/farmacología , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Fragmentos de Péptidos , Telomerasa/genética , Telomerasa/metabolismo , Telomerasa/farmacología
11.
Aging (Albany NY) ; 13(3): 3202-3217, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33539321

RESUMEN

Prostate cell proliferation, driven by testosterone, is a major characteristic of benign prostatic hyperplasia (BPH). GV1001, a human telomerase reverse transcriptase catalytic subunit, is an injectable formulation used as a cancer vaccine. It functions as a cell penetrating peptide to regulate cell proliferation. Here, we found that GV1001 effectively suppressed proliferation of prostatic stromal myofibroblasts (WPMY-1) and prostatic epithelial cells (RWPE-1 and WPE-NA22) treated with dihydrotestosterone. Also, GV1001 bound to androgen receptors (ARs) in the cytosol of stromal and epithelial cells. In an experimental animal model implanted with an infusion pump for spontaneous and continuous release of testosterone, revealed that GV1001 reduced prostatic hypertrophy and inhibited the cell proliferation and the expression of Ki67, proliferating cell nuclear antigen, and prostate specific antigen. In addition, GV1001 prevented fibrosis of the prostate by downregulating expression of prostatic epithelial-mesenchymal transition (EMT)-related proteins such as transforming growth factor (TGF)-ß, Snail, Slug, N-cadherin, and Vimentin, and by up-regulating E-cadherin. Taken together, these results suggest that GV1001, which suppresses TGF-ß-mediated EMT by outcompeting testosterone for binding to AR, is a potential therapeutic drug for BPH accompanied by prostatic fibrosis.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Hiperplasia Prostática/metabolismo , Receptores Androgénicos , Telomerasa/farmacología , Animales , Línea Celular , Dihidrotestosterona/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores Androgénicos/efectos de los fármacos , Receptores Androgénicos/metabolismo
12.
Mol Ther ; 29(4): 1395-1410, 2021 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-33388418

RESUMEN

Doxorubicin is one of the most potent chemotherapeutic agents. However, its clinical use is restricted due to the severe risk of cardiotoxicity, partially attributed to elevated production of reactive oxygen species (ROS). Telomerase canonically maintains telomeres during cell division but is silenced in adult hearts. In non-dividing cells such as cardiomyocytes, telomerase confers pro-survival traits, likely owing to the detoxification of ROS. Therefore, we hypothesized that pharmacological overexpression of telomerase may be used as a therapeutic strategy for the prevention of doxorubicin-induced cardiotoxicity. We used adeno-associated virus (AAV)-mediated gene therapy for long-term expression of telomerase in in vitro and in vivo models of doxorubicin-induced cardiotoxicity. Overexpression of telomerase protected the heart from doxorubicin-mediated apoptosis and rescued cardiac function, which was accompanied by preserved cardiomyocyte size. At the mechanistic level, we observed altered mitochondrial morphology and dynamics in response to telomerase expression. Complementary in vitro experiments confirmed the anti-apoptotic effects of telomerase overexpression in human induced pluripotent stem cell-derived cardiomyocytes after doxorubicin treatment. Strikingly, elevated levels of telomerase translocated to the mitochondria upon doxorubicin treatment, which helped to maintain mitochondrial function. Thus, telomerase gene therapy could be a novel preventive strategy for cardiotoxicity by chemotherapy agents such as the anthracyclines.


Asunto(s)
Cardiotoxicidad/genética , Doxorrubicina/efectos adversos , Neoplasias/tratamiento farmacológico , Telomerasa/genética , Animales , Apoptosis/efectos de los fármacos , Cardiotoxicidad/prevención & control , Cardiotoxicidad/terapia , Dependovirus/genética , Doxorrubicina/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Vectores Genéticos/genética , Vectores Genéticos/farmacología , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Miocitos Cardíacos/efectos de los fármacos , Neoplasias/complicaciones , Neoplasias/genética , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Telomerasa/farmacología
13.
Front Immunol ; 11: 652, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32508804

RESUMEN

Previously, a telomerase-derived 16-mer peptide, GV1001, developed as an anticancer vaccine, was reported to exert antiviral effects on human immunodeficiency virus or hepatitis C virus in a heat shock protein-dependent manner. Here we investigated whether GV1001 exerts antiviral effects on hepatitis B virus (HBV) and elucidated its underlying mechanisms. GV1001 inhibited HBV replication and hepatitis B surface antigen (HBsAg) secretion in a dose-dependent manner, showing synergistic antiviral effects with nucleos(t)ide analogs (NAs) including entecavir and lamivudine. This peptide also inhibited viral cccDNA and pgRNA. The intravenous GV1001 treatment of transgenic mice had anti-HBV effects. Our mechanistic studies revealed that GV1001 suppresses HBV replication by inhibiting capsid formation via type I interferon-mediated induction of heme oxygenase-1 (HO-1). GV1001 promoted the mitochondrial DNA stress-mediated release of oxidized DNA into the cytosol, resulting in IFN-I-dependent anti-HBV effects via the STING-IRF3 axis. We found that the anti-HBV effect of GV1001 was due to its ability to penetrate into the cytosol via extracellular heat shock protein, leading to phagosomal escape-mediated mtDNA stress. We demonstrated that the cell-penetrating and cytosolic localization capacity of GV1001 results in antiviral effects on HBV infections via mtDNA stress-mediated IFN-I production. Thus, GV1001, a peptide proven to be safe for human use, may be an anti-HBV drug that can be synergistically used with nucleot(s)ide analog.


Asunto(s)
Antivirales/metabolismo , Daño del ADN/inmunología , ADN Mitocondrial/genética , Virus de la Hepatitis B/fisiología , Hepatitis B/inmunología , Interferón Tipo I/metabolismo , Fragmentos de Péptidos/farmacología , Telomerasa/farmacología , Animales , Sinergismo Farmacológico , Guanina/análogos & derivados , Guanina/farmacología , Hemo-Oxigenasa 1/metabolismo , Células Hep G2 , Hepatitis B/tratamiento farmacológico , Antígenos de Superficie de la Hepatitis B/metabolismo , Humanos , Factor 3 Regulador del Interferón/metabolismo , Lamivudine/farmacología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Fragmentos de Péptidos/uso terapéutico , Fagosomas/metabolismo , Transducción de Señal , Telomerasa/uso terapéutico , Replicación Viral
14.
Endocr Relat Cancer ; 26(2): 147-162, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30400054

RESUMEN

GV1001, a 16-amino acid fragment of the human telomerase reverse transcriptase catalytic subunit (hTERT), has been developed as an injectable formulation of cancer vaccine. Here, we revealed for the first time that GV1001 is a novel ligand for gonadotropin-releasing hormone receptor (GnRHR). The docking prediction for GV1001 against GnRHR showed high binding affinity. Binding of GV1001 to GnRHR stimulated the Gαs-coupled cAMP signaling pathway and antagonized Gαq-coupled Ca2+ release by leuprolide acetate (LA), a GnRHR agonist. Repeated injection of GV1001 attenuated both serum testosterone level and seminal vesicle weight via desensitization of hypothalamic-pituitary-gonadal (HPG) axis. We then tested whether GV1001 has an inhibitory effect on tumor growth of LNCaP cells, androgen receptor-positive human prostate cancer (PCa) cells. GV1001 significantly inhibited tumor growth and induced apoptosis in LNCaP-implanted xenografts. Interestingly, mRNA expressions of matrix metalloproteinase 2 and matrix metalloproteinase 9 were suppressed by GV1001, but not by LA. Moreover, GV1001 significantly inhibited the proliferation and migration of PCa cells and induced apoptosis in a concentration-dependent manner. Our findings suggest that GV1001 functions as a biased GnRHR ligand to selectively stimulate the Gαs/cAMP pathway, with anti-proliferative and anti-migratory effects on human PCa.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Fragmentos de Péptidos/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Receptores LHRH/genética , Telomerasa/uso terapéutico , Animales , Vacunas contra el Cáncer/farmacología , Humanos , Ligandos , Masculino , Ratones , Fragmentos de Péptidos/farmacología , Neoplasias de la Próstata/patología , Transducción de Señal , Telomerasa/farmacología
15.
Int J Mol Med ; 42(4): 2260-2268, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30015834

RESUMEN

Benign prostatic hyperplasia (BPH) is one of the leading causes of male reproductive disorders. Therapeutic agents currently in use have severe side effects; therefore, alternative drugs that exhibit improved therapeutic activity without side effects are required. The present study investigated the protective effect of GV1001 against testosterone­induced BPH in rats. BPH in castrated rats was established via daily subcutaneous (s.c.) injections of testosterone propionate (TP, 3 mg/kg) dissolved in corn oil for 4 weeks. GV1001 (0.01, 0.1 and 1 mg/kg, s.c.) was administered 3 times per week for 4 weeks, together with TP (3 mg/kg) injection. The rats were sacrificed on the last day of treatment, and their prostates were excised and weighed for biochemical and histological studies. Serum levels of testosterone and dihydrotestosterone (DHT) were also measured. In rats with TP­induced BPH, a significant increase in prostate weight (PW) and prostatic index (PI), accompanied by a decrease in antioxidant enzyme activity, was observed. Histological studies revealed clearly enlarged glandular cavities in rats with BPH. GV1001 (0.01 and 0.1 mg/kg) treatment significantly decreased PW and PI in rats with TP­induced BPH. In addition, GV1001 demonstrated a potent inhibitory effect on 5α­reductase in prostate. The present data suggest that the protective role of GV1001 against testosterone­induced BPH is closely associated with its antioxidant potential. Additional studies are required to identify the mechanisms by which GV1001 protects against BPH to determine its clinical application.


Asunto(s)
Fragmentos de Péptidos/farmacología , Hiperplasia Prostática/tratamiento farmacológico , Telomerasa/farmacología , Testosterona/efectos adversos , Animales , Modelos Animales de Enfermedad , Masculino , Hiperplasia Prostática/inducido químicamente , Hiperplasia Prostática/metabolismo , Hiperplasia Prostática/patología , Ratas , Ratas Sprague-Dawley , Testosterona/farmacología
16.
Int J Mol Med ; 41(6): 3211-3220, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29568955

RESUMEN

GV1001 is a 16­amino acid peptide derived from the human telomerase reverse transcriptase (hTERT) protein (616­626; EARPALLTSRLRFIPK), which lies within the reverse transcriptase domain. Originally developed as an anticancer vaccine, GV1001 demonstrates diverse cellular effects, including anti­inflammatory, tumor suppressive and antiviral effects. In the present study, the radioprotective and antifibrotic effects of GV1001 were demonstrated through suppressing transforming growth factor­ß (TGF­ß) signaling. Proliferating human keratinocytes underwent premature senescence upon exposure to ionizing radiation (IR), however, treatment of cells with GV1001 allowed the cells to proliferate and showed a reduction in senescent phenotype. GV1001 treatment notably increased the levels of Grainyhead­like 2 and phosphorylated (p­)Akt (Ser473), and reduced the activation of p53 and the level of p21/WAF1 in irradiated keratinocytes. It also markedly suppressed the level of TGF­ß signaling molecules, including p­small mothers against decapentaplegic (Smad)2/3 and Smad4, and TGF­ß target genes, including zinc finger E­box binding homeobox 1, fibronectin, N­cadharin and Snail, in irradiated keratinocytes. Furthermore, GV1001 suppressed TGF­ß signaling in primary human fibroblasts and inhibited myofibroblast differentiation. Chromatin immunoprecipitation revealed that GV1001 suppressed the binding of Smad2 on the promoter regions of collagen type III α1 chain (Col3a1) and Col1a1. In a dermal fibrosis model in vivo, GV1001 treatment notably reduced the thickness of fibrotic lesions and the synthesis of Col3a1. These data indicated that GV1001 ameliorated the IR­induced senescence phenotype and tissue fibrosis by inhibiting TGF­ß signaling and may have therapeutic effects on radiation­induced tissue damage.


Asunto(s)
Fragmentos de Péptidos/química , Fragmentos de Péptidos/farmacología , Telomerasa/química , Telomerasa/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Inmunoprecipitación de Cromatina , Humanos , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Cicatrización de Heridas/efectos de los fármacos
17.
Liver Int ; 38(9): 1635-1645, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29405561

RESUMEN

BACKGROUND AND AIMS: Human telomerase reverse transcriptase is a catalytic enzyme involved in telomere elongation. It is expressed in many tumours, including hepatocellular carcinoma. The purpose of the present study was to identify major histocompatibility complex class II-restricted helper T cell epitopes derived from human telomerase reverse transcriptase in patients with hepatocellular carcinoma. METHODS: TEPITOPE software was used to predict helper T cell epitopes based on the entire amino acid sequence of human telomerase reverse transcriptase, and peptides were synthesized based on the predicted sequence. Interferon (IFN)-γ enzyme linked immunospot assay was performed to examine the T cell response to each of the synthesized peptides in peripheral blood mononuclear cells. Furthermore, the peptides were labelled with fluorescein isothiocyanate to test their binding affinity for major histocompatibility complex class II molecules. Lastly, the association between patient characteristics and the level of immune response to these epitopes was examined. RESULTS: Positive T cell response (>10% enzyme linked immunospot positivity) was detected against 4 of 10 peptides. Among all peptides, positive T cell response to the hTERT68 peptide was detected most frequently. While hTERT68 was HLA-DRB1*0405-restricted, it also bound to other MCH class II molecules. Positive helper T cell response was detected most frequently in hepatocellular carcinoma patients with a low serum alpha-foetoprotein level. Several treatments for hepatocellular carcinoma enhanced the immune response against the peptides. CONCLUSION: Our findings indicate that helper T cell epitopes identified in the present study may be useful to investigate immune responses and for immunotherapy in hepatocellular carcinoma patients.


Asunto(s)
Carcinoma Hepatocelular/inmunología , Antígenos HLA-DR/química , Neoplasias Hepáticas/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Telomerasa/inmunología , Anciano , Secuencia de Aminoácidos , Carcinoma Hepatocelular/terapia , Estudios de Casos y Controles , Ensayo de Immunospot Ligado a Enzimas , Epítopos de Linfocito T/química , Femenino , Citometría de Flujo , Humanos , Inmunoterapia , Japón , Neoplasias Hepáticas/terapia , Masculino , Persona de Mediana Edad , Péptidos/metabolismo , Telomerasa/farmacología
18.
Elife ; 72018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29378675

RESUMEN

Pulmonary fibrosis is a fatal lung disease characterized by fibrotic foci and inflammatory infiltrates. Short telomeres can impair tissue regeneration and are found both in hereditary and sporadic cases. We show here that telomerase expression using AAV9 vectors shows therapeutic effects in a mouse model of pulmonary fibrosis owing to a low-dose bleomycin insult and short telomeres. AAV9 preferentially targets regenerative alveolar type II cells (ATII). AAV9-Tert-treated mice show improved lung function and lower inflammation and fibrosis at 1-3 weeks after viral treatment, and improvement or disappearance of the fibrosis at 8 weeks after treatment. AAV9-Tert treatment leads to longer telomeres and increased proliferation of ATII cells, as well as lower DNA damage, apoptosis, and senescence. Transcriptome analysis of ATII cells confirms downregulation of fibrosis and inflammation pathways. We provide a proof-of-principle that telomerase activation may represent an effective treatment for pulmonary fibrosis provoked or associated with short telomeres.


Asunto(s)
Terapia Genética/métodos , Fibrosis Pulmonar/terapia , Telomerasa/farmacología , Telómero/metabolismo , Células Epiteliales Alveolares/fisiología , Animales , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Pulmón/patología , Pulmón/fisiología , Ratones , Fibrosis Pulmonar/patología , Pruebas de Función Respiratoria , Telomerasa/genética , Usos Terapéuticos
19.
Urology ; 113: 129-137, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29154986

RESUMEN

OBJECTIVE: To investigate the anticancer effects of GV1001 and its biological mechanism of action in renal cell carcinoma (RCC). METHODS: The effects of GV1001 on cell survival and apoptosis in RCC cells were examined in vitro using cell viability assay, fluorescence-activated cell sorting, and terminal deoxynucleotidyl transferase dUTP nick end labeling assay. To evaluate the effect of GV1001 on migration, invasion, and angiogenesis, we used wound healing, invasion, endothelial cell tube formation assay, and western blot analysis. Furthermore, we used an RCC xenograft model with either phosphate buffered saline or GV1001 to confirm the anticancer effect of GV1001 in vivo. Tumor volume was monitored during treatment, and tumor weight was measured after animals were killed. Apoptosis and angiogenesis of the tumor tissue were assessed using hematoxylin and eosin staining, immunohistochemistry, and western blot analysis. RESULTS: GV1001 reduced cell viability and induced apoptosis in RCC cells in vitro. Furthermore, GV1001 suppressed the migration and invasion of RCC cells through regulation of matrix metalloproteinases and tissue inhibitors of metalloproteinases. In addition, GV1001 reduced angiogenesis via regulation of hypoxia-inducible factor 1α. In xenograft mouse model experiment, GV1001 reduced tumor growth and induced apoptosis. As in the in vitro results, GV1001 significantly reduced angiogenesis through regulation of hypoxia-inducible factor 1α in vivo. CONCLUSION: Our data demonstrated that GV1001 induced apoptosis through suppression of angiogenesis in RCCs both in vitro and in vivo, which suggests that GV1001 may be a potential therapeutic target for RCC.


Asunto(s)
Apoptosis/efectos de los fármacos , Carcinoma de Células Renales/tratamiento farmacológico , Neoplasias Renales/tratamiento farmacológico , Neoplasias Experimentales/tratamiento farmacológico , Neovascularización Patológica/prevención & control , Fragmentos de Péptidos/farmacología , Telomerasa/farmacología , Animales , Antineoplásicos/farmacología , Carcinoma de Células Renales/patología , Línea Celular Tumoral/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Xenoinjertos , Humanos , Etiquetado Corte-Fin in Situ , Técnicas In Vitro , Neoplasias Renales/patología , Ratones , Sensibilidad y Especificidad , Resultado del Tratamiento
20.
Mol Med Rep ; 16(5): 7315-7320, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28944828

RESUMEN

The potential cardioprotective effects of the novel vaccine peptide GV1001 were evaluated in myocardial ischemia­reperfusion injury induced rat models. GV1001 is a human telomerase reverse transcriptase derived peptide, which has been reported to possess both anti­tumor and anti­inflammatory effects. The normal saline (control group) and various concentrations (0.001­10 mg/kg) of GV1001 were administered directly to the right ventricle anterior wall before induction of ischemia. The was induced by Tightening the snare around the left anterior descending coronary artery for 40 min, before releasing the snare for 10 min induced the myocardial ischemia­reperfusion injury and was conducted in Sprague­Dawley rats. The area at risk, histology, apoptotic cells, neutrophils and inflammatory cytokines were analyzed from the excised heart tissue following myocardial ischemia­reperfusion injury. The area at risk was protected by concentrations of GV1001 equal to or higher than 0.01 mg/kg. At 0.1 mg/kg and higher concentrations of GV1001, the hemorrhage in the heart was attenuated, while severe congestion was reported in the control group. Apoptotic cells, myeloperoxidase activity and inflammatory cytokines [tumor necrosis factor (TNF)­α and interleukin (IL)­6] revealed decreased levels in a dose­dependent manner with respect to GV1001 concentration. The group treated with 10 mg/kg GV1001 demonstrated 59.73% apoptotic cells (P<0.001), 48.14% neutrophil contents (P<0.001), 55.63% TNF­α (P<0.01) and 42.35% IL­6 (P<0.01) levels, compared with the control group. The novel vaccine peptide GV1001 provided protective effects on myocardial ischemia­reperfusion injury and, therefore, it should be considered as an alternative potential anti­inflammatory agent for myocardial ischemia­reperfusion injury.


Asunto(s)
Daño por Reperfusión Miocárdica/prevención & control , Fragmentos de Péptidos/uso terapéutico , Sustancias Protectoras/uso terapéutico , Telomerasa/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Hiperemia/etiología , Interleucina-6/análisis , Interleucina-6/metabolismo , Masculino , Daño por Reperfusión Miocárdica/patología , Miocardio/metabolismo , Miocardio/patología , Neutrófilos/citología , Fragmentos de Péptidos/efectos adversos , Fragmentos de Péptidos/farmacología , Peroxidasa/metabolismo , Sustancias Protectoras/efectos adversos , Sustancias Protectoras/farmacología , Ratas , Ratas Sprague-Dawley , Telomerasa/efectos adversos , Telomerasa/farmacología , Factor de Necrosis Tumoral alfa/análisis , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...