1
|
Rajkumar RP. Telomere Dynamics in Post-Traumatic Stress Disorder: A Critical Synthesis. Biomedicines 2025; 13:507. [PMID: 40002919 PMCID: PMC11853385 DOI: 10.3390/biomedicines13020507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 01/03/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Post-traumatic stress disorder (PTSD), a mental disorder caused by exposure to traumatic stress, affects 5-10% of the world's population. There is some evidence that PTSD is associated with accelerated cellular aging, leading to an increased risk of medical and neurodegenerative comorbidities. Alterations in telomere length (TL) and telomerase enzyme activity have been proposed as biomarkers of this process. This hypothesis was seemingly confirmed in preliminary research, but more recent studies have yielded mixed results. The current narrative review was conducted to provide a critical synthesis of existing research on telomere length and telomerase in PTSD. Data from 26 clinical studies suggest that TL in PTSD is highly variable and may be influenced by methodological, demographic, trauma-related, and psychosocial factors. There is no evidence for altered telomerase activity in PTSD. In contrast, animal research suggests that exposure to traumatic stress does lead to TL shortening. Overall, it is likely that TL is not, by itself, a reliable biomarker of cellular aging in PTSD. Other markers of cellular senescence, such as epigenetic changes, may prove to be more specific in measuring this process in patients with PTSD.
Collapse
Affiliation(s)
- Ravi Philip Rajkumar
- Department of Psychiatry, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry 605006, India
| |
Collapse
|
2
|
Dou J, Hu W, Zhang X, Jiang K. NOP10 predicts poor prognosis and promotes pancreatic cancer progression. BMC Cancer 2024; 24:1394. [PMID: 39538226 PMCID: PMC11558815 DOI: 10.1186/s12885-024-13180-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 11/11/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Telomere shortening and RNA pseudo-uridylation are common features of tumors. NOP10 is a member of the H/ACA snoRNP family, essential for maintaining telomerase activity and RNA pseudouridylation. NOP10 has been indicated to be substantially expressed in tumors such as breast and lung cancers and is associated with poor prognosis. Currently, no investigation exists on NOP10 in pancreatic cancer (PC). This is the first investigation to elucidate the impact on tumorigenesis and prognostic value of NOP10 in pancreatic adenocarcinoma (PAAD). METHOD NOP10 expression and its survival prognostic significance were analyzed via clinical PAAD data from the TCGA database and NOP10 expression in other tumors from the GEPIA database. Furthermore, the NOP10 expression and survival prognosis in clinical samples were validated by qRT-PCR. In-vitro experiments were carried out to elucidate the impact of NOP10 on the biological function of PC cells. RESULTS It was revealed that NOP10 expression was increased in PC tissues than in the normal pancreatic tissues. High NOP10 expression was markedly linked with poorer prognosis. NOP10 may be involved in focal adhesion, channel activity, cAMP signaling pathway, the interaction of neuroactive ligand-receptor, and cell adhesion molecules cams. NOP10 was associated with the tumour immune microenvironment and drug sensitivity. Down-regulation of NOP10 expression suppressed PC cells' ability to proliferate, migrate, and invade. CONCLUSIONS This investigation elucidated the prognostic and predictive importance of NOP10 in PAAD and revealed that NOP10 is associated with poor prognostic features, survival prognosis and TIME. Knockdown of NOP10 inhibits the progression of PAAD.
Collapse
Affiliation(s)
- Jin Dou
- Medical College, Yangzhou University, Yangzhou, China
- Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| | - Weikang Hu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaoyu Zhang
- Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China.
| | - Kuirong Jiang
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
3
|
de Jaeger C, Kruiskamp S, Voronska E, Lamberti C, Baramki H, Beaudeux JL, Cherin P. A Natural Astragalus-Based Nutritional Supplement Lengthens Telomeres in a Middle-Aged Population: A Randomized, Double-Blind, Placebo-Controlled Study. Nutrients 2024; 16:2963. [PMID: 39275278 PMCID: PMC11397652 DOI: 10.3390/nu16172963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024] Open
Abstract
Telomeres are ribonucleoprotein structures that form a protective buffer at the ends of chromosomes, maintaining genomic integrity during the cell cycle. A decrease in average telomere length is associated with with age and with aging-related diseases such as cancer and cardiovascular disease. In this study, we conducted a randomized, double-blind, placebo-controlled trial over six months to compare the effects of the Astragalus-based supplement versus a placebo on telomere length (TL) in 40 healthy volunteers (mean age 56.1 ± 6.0 years). Twenty subjects received the supplement, and 20 received placebo capsules. All participants completed the study, and no adverse side effects were reported at six months. Subjects taking the Astragalus-based supplement exhibited significantly longer median TL (p = 0.01) and short TL (p = 0.004), along with a lower percentage of short telomeres, over the six-month period, while the placebo group showed no change in TL. This trial confirmed that the supplement significantly lengthens both median and short telomeres by increasing telomerase activity and reducing the percentage of short telomeres (<3 Kbp) in a statistically and possibly clinically significant manner. These results align with a previous open prospective trial, which found no toxicity associated with the supplement's intake. These findings suggest that this Astragalus-based supplement warrants further investigation for its potential benefits in promoting health, extending life expectancy, and supporting healthy aging.
Collapse
Affiliation(s)
- Christophe de Jaeger
- Institute of Medicine and Physiology of Longevity (Institut de Jaeger), 127, rue de la Faisanderie, 75016 Paris, France; (S.K.); (E.V.); (C.L.); (H.B.)
| | - Saskia Kruiskamp
- Institute of Medicine and Physiology of Longevity (Institut de Jaeger), 127, rue de la Faisanderie, 75016 Paris, France; (S.K.); (E.V.); (C.L.); (H.B.)
| | - Elena Voronska
- Institute of Medicine and Physiology of Longevity (Institut de Jaeger), 127, rue de la Faisanderie, 75016 Paris, France; (S.K.); (E.V.); (C.L.); (H.B.)
| | - Carla Lamberti
- Institute of Medicine and Physiology of Longevity (Institut de Jaeger), 127, rue de la Faisanderie, 75016 Paris, France; (S.K.); (E.V.); (C.L.); (H.B.)
| | - Hani Baramki
- Institute of Medicine and Physiology of Longevity (Institut de Jaeger), 127, rue de la Faisanderie, 75016 Paris, France; (S.K.); (E.V.); (C.L.); (H.B.)
| | - Jean Louis Beaudeux
- Service de Biochimie Générale, Université Paris Cité, Assistance Publique-Hôpitaux de Paris, CHU Necker, Inserm UMR S_1139, 75015 Paris, France;
| | - Patrick Cherin
- Service de Médecine Interne 2, Institut E3M, Sorbonne Université, Assistance Publique-Hôpitaux de Paris, CHU Pitié-Salpêtrière, French National, Referral Center for Autoimmune Disorders, Inserm UMRS, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013 Paris, France;
| |
Collapse
|
4
|
Wang X, Deng H, Lin J, Zhang K, Ni J, Li L, Fan G. Distinct roles of telomerase activity in age-related chronic diseases: An update literature review. Biomed Pharmacother 2023; 167:115553. [PMID: 37738798 DOI: 10.1016/j.biopha.2023.115553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 09/24/2023] Open
Abstract
Although telomerase has low activity in somatic quiescent cells, it plays an significant roles in regenerative cells such as endothelial cells, hepatocytes, epithelial cells, and hemocytes. Telomerase activity and telomere length are critical factors in age-related chronic diseases as they are closely related to cell senescence. However, whether telomerase activity plays a key role in disease progression or whether the role of telomerase is unified among different diseases are unresolved. Considering that aging is the most important risk factor for neurodegenerative and metabolic diseases, this article will analyze the evidence, mechanism, and therapeutic potential of telomerase activity in several chronic disease, including type 2 diabetes, neurodegenerative diseases, atherosclerosis, heart failure and non-alcoholic fatty liver disease, in order to provide clues for the use of telomerase activity to target the treatment of age-related chronic diseases.
Collapse
Affiliation(s)
- Xiaodan Wang
- Medical Experiment Center, Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 300381 Tianjin, China
| | - Hao Deng
- Medical Experiment Center, Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 300381 Tianjin, China
| | - Jingyi Lin
- Medical Experiment Center, Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 300381 Tianjin, China
| | - Kai Zhang
- Medical Experiment Center, Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 300381 Tianjin, China
| | - Jingyu Ni
- Medical Experiment Center, Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 300381 Tianjin, China
| | - Lan Li
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae for the Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Guanwei Fan
- Medical Experiment Center, Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 300381 Tianjin, China.
| |
Collapse
|
5
|
Dou W, Xie J, Chen J, Zhou J, Xu Z, Wang Z, Zhu Q. Overexpression of adrenomedullin (ADM) alleviates the senescence of human dental pulp stem cells by regulating the miR-152/CCNA2 pathway. Cell Cycle 2023; 22:565-579. [PMID: 36310381 PMCID: PMC9928452 DOI: 10.1080/15384101.2022.2135621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
The limitation of human dental pulp stem cells (DPSCs), which have potential application value in regenerative medicine, is that they are prone to age in vitro. Studies have shown adrenomedullin (ADM) is believed to promote the proliferation of human DPSCs, but whether it can also affect aging remains to be investigated. A lentivirus vector was used to construct human DPSCs overexpressing ADM. Senescence tests were carried out on cells of the 7th and 15th passage. Transcriptome analysis was conducted to analyze microRNA expression regulation changes after human DPSCs overexpressed ADM. H2O2 induced the aging model of human DPSCs, and we examined the mechanism of recovery of aging through transfection experiments with miR-152 mimic, pCDH-CCNA2, and CCNA2 siRNA. Overexpression of ADM significantly upregulated the G2/M phase ratio of human DPSCs in natural passage culture (P = 0.001) and inhibited the expression of p53 (P = 0.014), P21 WAF1 (P = 0.015), and P16 INK4A (P = 0.001). Decreased ROS accumulation was observed in human DPSCs during long-term natural passage (P = 0.022). Transcriptome analysis showed that miR-152 was significantly upregulated during human DPSC senescence (P = 0.001) and could induce cell senescence by directly targeting CCNA2. Transfection with miR-152 mimic significantly reversed the inhibitory effect of ADM overexpression on p53 (P = 0.006), P21 WAF1 (P = 0.012), and P16 INK4A (P = 0.01) proteins in human DPSCs (H2O2-induced). In contrast, pCDH-CCNA2 weakened the effect of the miR-152 mimic, thus promoting cell proliferation and antiaging. ADM-overexpressing human DPSCs promote cell cycle progression and resist cellular senescence through CCNA2 expression promotion by inhibiting miR-152.
Collapse
Affiliation(s)
- Wenxue Dou
- Department of Stomatology, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai, China
| | - Jiaye Xie
- Department of Stomatology, Tongren Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jianan Chen
- Department of Stomatology, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai, China
| | - Jiajun Zhou
- Department of Stomatology, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Zunyue Xu
- Department of Stomatology, Shanghai Songjiang District Central Hospital, Shanghai, China
| | - Zheng Wang
- Department of Stomatology, Tongren Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qiang Zhu
- Department of Stomatology, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai, China,CONTACT Qiang Zhu Department of Stomatology, The First Affiliated Hospital of Naval Medical University: Changhai Hospital, Shanghai200433, China
| |
Collapse
|
6
|
Heat-Killed Staphylococcus aureus Induces Bone Mass Loss through Telomere Erosion. Int J Mol Sci 2023; 24:ijms24043179. [PMID: 36834587 PMCID: PMC9960843 DOI: 10.3390/ijms24043179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/30/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023] Open
Abstract
The mechanism of systemic osteoporosis caused by chronic infection is not completely clear, and there is a lack of reasonable interventions for this disease. In this study, heat-killed S. aureus (HKSA) was applied to simulate the inflammation caused by the typical clinical pathogen and to explore the mechanism of systemic bone loss caused by it. In this study, we found that the systemic application of HKSA caused bone loss in mice. Further exploration found that HKSA caused cellular senescence, telomere length shortening, and telomere dysfunction-induced foci (TIF) in limb bones. As a well-known telomerase activator, cycloastragenol (CAG) significantly alleviated HKSA-induced telomere erosion and bone loss. These results suggested that telomere erosion in bone marrow cells is a possible mechanism of HKSA-induced bone loss. CAG may protect against HKSA-induced bone loss by alleviating telomere erosion in bone marrow cells.
Collapse
|
7
|
Wu Y, Zhang Y, Jiao J. The relationship between n-3 polyunsaturated fatty acids and telomere: A review on proposed nutritional treatment against metabolic syndrome and potential signaling pathways. Crit Rev Food Sci Nutr 2022; 64:4457-4476. [PMID: 36330807 DOI: 10.1080/10408398.2022.2142196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Metabolic syndrome (MetS), a cluster of metabolic abnormalities composed of central obesity, elevated blood pressure, glucose disturbances, hypercholesterolemia and dyslipidaemia, has increasingly become a public health problem in the 21st century worldwide. The dysfunction of telomeres, the repetitive DNA with highly conserved sequences (5'-TTAGGG-3'), is remarkably correlated with organismal aging, even suggesting a causal relationship with metabolic disorders. The health benefits of n-3 polyunsaturated fatty acids (PUFAs) in multiple disorders are associated with telomere length in evidence, which have recently drawn wide attention. However, functional targets and pathways for the associations of n-3 PUFAs and telomere with MetS remain scare. Few studies have summarized the role of n-3 PUFAs in DNA damage repair pathways, anti-inflammatory pathways, and redox balance, linking with telomere biology, and other potential telomere-related signaling pathways. This review aims to (i) elucidate how n-3 PUFAs ameliorate telomere attrition in the context of anti-oxidation and anti-inflammation; (ii) unravel the role of n-3 PUFAs in modulating telomere-related neuron dysfunction and regulating the neuro-endocrine-immunological network in MetS; (iii) epidemiologically implicate the associations of metabolic disorders and n-3 PUFAs with telomere length; and (iv) suggest promising biochemical approaches and advancing methodologies to overcome the inter-variation problem helpful for future research.
Collapse
Affiliation(s)
- Yuqi Wu
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yu Zhang
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jingjing Jiao
- Department of Nutrition, School of Public Health, Department of Clinical Nutrition, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
8
|
Rad54L promotes bladder cancer progression by regulating cell cycle and cell senescence. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:185. [PMID: 36071250 DOI: 10.1007/s12032-022-01751-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 05/13/2022] [Indexed: 10/14/2022]
Abstract
Bladder cancer (BCa) is the most prevalent cancer of the urinary system, but its pathogenesis is still poorly understood. Several reports have suggested that gene damage repair is highly correlated with tumor development and drug resistance, in which homologous recombination repair gene Rad54L seems to play an important role, through yet unclear mechanisms. Therefore, this study stratified cancer patients by Rad54L expression in BCa tissue, and high Rad54L expression was associated with a poor prognosis. Mechanistically, we demonstrate that high Rad54L expression promotes abnormal bladder tumor cell proliferation by changing the cell cycle and cell senescence. In addition, this study also suggests that Rad54L may be associated with p53, p21, and pRB in BCa tissue. In summary, this study exposes Rad54L as potential a prognostic biomarker and precision treatment target in BCa.
Collapse
|
9
|
Tade RS, Patil PO. Biofabricated functionalized graphene quantum dots (fGQDs): Unravelling its fluorescence sensing mechanism of human telomerase reverse transcriptase (hTERT) antigen and in vitro bioimaging application. Biomed Mater 2022; 17. [PMID: 35896107 DOI: 10.1088/1748-605x/ac84ba] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 07/27/2022] [Indexed: 11/11/2022]
Abstract
Lung cancer (LC) is a deadly malignancy that is posing a serious threat to human health. Therefore, early detection of LC biomarkers is the key to reducing LC-related fatalities. Herein, we present the first fluorescent-based selective detection of LC biomarker human telomerase reverse transcriptase (hTERT) using polyethyleneimine (PEI) functionalized graphene quantum dots (fGQDs). One-pot in situ synthesis of amine-functionalized GQDs was accomplished by hydrothermal carbonization of biowaste-derived cellulose and PEI. Synthesized fGQDs were characterized by various analytical techniques. Synthesized fGQDs not only exhibited enhanced fluorescence life-time but also excellent stability in the different solvents compared to bare GQDs. The surface activation of hTERT-Ab by carbodiimide chemistry (EDC-NHS) resulted in stacking interactions with fGQDs, involving adsorption-desorption as well as competitive mechanisms. The higher inherent affinity of hTERT-Ag (hTERT antigen) for hTERT-Ab (hTERT antibody) resulted in complex formation and recovery of fGQD fluorescence. As a result, this fluorescence sensing demonstrated a greater linear detection range (0.01 ng mL-1 to 100 µg mL-1) as well as a notable low detection limit (36.3 pg mL-1). Furthermore, the fabricated immunosensor (Ab@fGQDs) has excellent stability and performance in real samples, with an average recovery of 97.32%. The results of cytotoxicity and cellular bioimaging study in A549 cells show that fGQDs can be used for additional nanotherapeutics and biological applications.
Collapse
Affiliation(s)
- Rahul S Tade
- Pharmaceutics, HR Patel Institute of Pharmaceutical Education and Research, Shirsoli PB, Jalgaon, Shirpur, Maharashtra, 425405, INDIA
| | - Pravin O Patil
- Department of Pharmaceutical Chemistry, H R Patel Institute of Pharmaceutical Education and Research, Shirpur 425405, Shirpur, Shirpur, 425405, INDIA
| |
Collapse
|
10
|
KLF4 regulates TERT expression in alveolar epithelial cells in pulmonary fibrosis. Cell Death Dis 2022; 13:435. [PMID: 35508454 PMCID: PMC9068714 DOI: 10.1038/s41419-022-04886-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/14/2022] [Accepted: 04/25/2022] [Indexed: 12/14/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) was considered as a telomere-mediated disease. TERT and TERC correlated with telomere length. Although telomerase gene mutations were associated with IPF, majority patients did not carry mutations. The mechanism by which telomerase expression was regulated in IPF are still unclear. In this study, we aimed to delineate the mechanisms that how TERT protein expression were regulated in alveolar epithelial cells (AECs) in pulmonary fibrosis. Here, we found that P16, P21 and fibrosis markers (αSMA and Collagen-I) were prominently increased in lung tissues of IPF patients and bleomycin-induced mouse models, while the expression of KLF4 and TERT were decreased in AECs. In vivo experiments, AAV-6 vectors mediated KLF4 over-expression with specific SP-C promoter was constructed. Over-expression of KLF4 in AECs could protect TERT expression and suppress the development of pulmonary fibrosis in bleomycin-induced mouse models. In the mechanism exploration of TERT regulation, KLF4 and TERT were both down-regulated in bleomycin-induced senescent MLE-12 and BEAS-2B cells. Compared with control group, small-interfering RNA targeting KLF4 significantly reduced the TERT expression and telomerase activity, while overexpression of KLF4 can increased the expression of TERT and telomerase activity in senescent AECs. Furthermore, ChIP showed that KLF4 protein could bind to the TERT promoter region in MLE-12 cells, suggesting that KLF4 could implicate in pathogenesis of lung fibrosis through regulating TERT transcription in AECs. Taken together, this study identified that KLF4 might be a promising potential target for further understanding the mechanism and developing novel strategy for the treatment of lung fibrosis in IPF.
Collapse
|
11
|
Methylation of Subtelomeric Chromatin Modifies the Expression of the lncRNA TERRA, Disturbing Telomere Homeostasis. Int J Mol Sci 2022; 23:ijms23063271. [PMID: 35328692 PMCID: PMC8955364 DOI: 10.3390/ijms23063271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/08/2022] [Accepted: 03/10/2022] [Indexed: 02/01/2023] Open
Abstract
The long noncoding RNA (lncRNA) telomeric repeat-containing RNA (TERRA) has been associated with telomeric homeostasis, telomerase recruitment, and the process of chromosome healing; nevertheless, the impact of this association has not been investigated during the carcinogenic process. Determining whether changes in TERRA expression are a cause or a consequence of cell transformation is a complex task because studies are usually carried out using either cancerous cells or tumor samples. To determine the role of this lncRNA in cellular aging and chromosome healing, we evaluated telomeric integrity and TERRA expression during the establishment of a clone of untransformed myeloid cells. We found that reduced expression of TERRA disturbed the telomeric homeostasis of certain loci, but the expression of the lncRNA was affected only when the methylation of subtelomeric bivalent chromatin domains was compromised. We conclude that the disruption in TERRA homeostasis is a consequence of cellular transformation and that changes in its expression profile can lead to telomeric and genomic instability.
Collapse
|
12
|
Greer PFC, Rich A, Coates DE. Effects of galectin-1 inhibitor OTX008 on oral squamous cell carcinoma cells in vitro and the role of AP-1 and the MAPK/ERK pathway. Arch Oral Biol 2021; 134:105335. [PMID: 34891102 DOI: 10.1016/j.archoralbio.2021.105335] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/17/2021] [Accepted: 11/29/2021] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To investigate the in vitro effects of inhibiting galectin-1 using the small-molecule inhibitor OTX008 on oral squamous cell carcinoma (OSCC) cell lines and the role of the MAPK pathway. METHODS One normal oral keratinocyte (NOK) and three OSCC cell lines were cultured in vitro and the expression of galectin-1 protein by each quantified using ELISA. Cell lines were treated with galectin-1 (50, 100 and 150 ng/mL) or OTX008 (12.5, 25, 50 and 100 μg/mL) and cell viability assayed (n = 3). OSCC cell lines with and without 25 μg/mL OTX008 (n = 3) treatment for 48 h, were analysed using qRT2-PCR with a custom array, to assess relative gene expression. RESULTS All cell lines were found to express galectin-1 protein. Exogenous galectin-1 significantly reduced cell viability in one OSCC cell line over time while the others were only minimally affected. OTX008 treatment reduced cell viability in a dose and time-dependent manner in all cell lines and this was associated with significant regulation of FOS gene expression in the OSCC cell lines. CONCLUSION OTX008 decreased the viability of OSCC and NOK cells in a dose-dependent manner. The significant regulation of FOS suggests OTX008 causes early induction of the MAPK pathway via the immediate response gene FOS as a subunit of the AP-1 complex.
Collapse
Affiliation(s)
- Philippa F C Greer
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| | - Alison Rich
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| | - Dawn E Coates
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
13
|
Roque CR, Sampaio LR, Ito MN, Pinto DV, Caminha JSR, Nunes PIG, Raposo RS, Santos FA, Windmöller CC, Crespo-Lopez ME, Alvarez-Leite JI, Oriá RB, Pinheiro RF. Methylmercury chronic exposure affects the expression of DNA single-strand break repair genes, induces oxidative stress, and chromosomal abnormalities in young dyslipidemic APOE knockout mice. Toxicology 2021; 464:152992. [PMID: 34670124 DOI: 10.1016/j.tox.2021.152992] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 10/01/2021] [Accepted: 10/12/2021] [Indexed: 12/31/2022]
Abstract
Mercury (Hg) is one of the most toxic environmental pollutants, especially when methylated, forming methylmercury (MeHg). MeHg affects DNA repair, increases oxidative stress, and predisposes to cancer. MeHg neurotoxicity is well-known, but recently MeHg-associated cardiovascular effects were recognized. This study evaluated circulating lipids, oxidative stress, and genotoxicity after MeHg-chronic exposure (20 mg/L in drinking water) in C57BL/6J wild-type and APOE knockout (ko) mice, the latter, being spontaneously dyslipidemic. Experimental mice were assigned to four groups: non-intoxicated and MeHg-intoxicated wild-type mice and non-intoxicated and MeHg-intoxicated APOE ko mice. Plasma levels of triglycerides, total cholesterol (TC), HDL, and LDL were analyzed. Liver lipid peroxidation and splenic gene expression of xeroderma pigmentosum complementation groups A, C, D, and G (XPA, XPC, XPD, and XPG), X-ray repair cross-complementing protein 1 (XRCC1), and telomerase reverse transcriptase (TERT) were measured. Fur Hg levels confirmed chronic MeHg intoxication. MeHg exposure raises TC levels both in wild-type and APOE ko mice. HDL and LDL-cholesterol levels were increased only in the MeHg-challenged APOE ko mice. MeHg increased liver lipid peroxidation, regardless of the genetic background. Unintoxicated APOE ko mice showed higher expression of TERT than all other groups. APOE deficiency increases XPA expression, regardless of MeHg intoxication. Furthermore, MeHg-intoxicated mice had more cytogenetic abnormalities, effect which was independent of APOE deficiency. More studies are needed to dissect the interactions between circulating lipids, MeHg intoxication, and DNA-repair pathways even at young age, interactions that likely play critical roles in cell senescence and the risk for chronic disorders later in life.
Collapse
Affiliation(s)
- Cássia R Roque
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and Institute of Biomedicine, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Letícia R Sampaio
- Cancer Cytogenomics Laboratory, Drug Research, and Development Center, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Mayumi N Ito
- Cancer Cytogenomics Laboratory, Drug Research, and Development Center, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Daniel V Pinto
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and Institute of Biomedicine, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Juan S R Caminha
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and Institute of Biomedicine, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Paulo I G Nunes
- Natural Products Laboratory, Biomedicine Center, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Ramon S Raposo
- Experimental Biology core, Health Sciences, University of Fortaleza, Fortaleza, CE, Brazil
| | - Flávia A Santos
- Natural Products Laboratory, Biomedicine Center, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Cláudia C Windmöller
- Laboratory of Atherosclerosis and Nutritional Biochemistry, Department of Biochemistry and Immunology, ICB, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Maria Elena Crespo-Lopez
- Laboratory of Molecular Pharmacology, Institute of Biological Sciences, Federal University of Para, Belém, PA, Brazil
| | - Jacqueline I Alvarez-Leite
- Laboratory of Atherosclerosis and Nutritional Biochemistry, Department of Biochemistry and Immunology, ICB, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Reinaldo B Oriá
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology and Institute of Biomedicine, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil.
| | - Ronald F Pinheiro
- Cancer Cytogenomics Laboratory, Drug Research, and Development Center, Federal University of Ceara, Fortaleza, CE, Brazil
| |
Collapse
|