Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Medicine (Baltimore) ; 103(24): e38533, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38875394

ABSTRACT

Physical examination data are used to indicate individual health status and organ health, and understanding which physical examination data are indicative of physiological aging is critical for health management and early intervention. There is a lack of research on physical examination data and telomere length. Therefore, the present study analyzed the association between blood telomere length and physical examination indices in healthy people of different ages to investigate the role and association of various organs/systems with physiological aging in the human body. The present study was a cross-sectional study. Sixteen physical examination indicators of different tissue and organ health status were selected and analyzed for trends in relation to actual age and telomere length (TL). The study included 632 individuals with a total of 11,766 data for 16 physical examination indicators. Age was linearly correlated with 11 indicators. Interestingly, telomere length was strongly correlated only with the renal indicators eGFR (P < .001), CYS-C (P < .001), and SCR (P < .001). The study established that renal aging or injury is a risk factor for Physical aging of the human body. Early identification and management are essential to healthcare.


Subject(s)
Aging , Biomarkers , Telomere , Humans , Cross-Sectional Studies , Male , Female , Middle Aged , Aging/genetics , Aging/physiology , Adult , Aged , Biomarkers/blood , Young Adult , Physical Examination/methods , Aged, 80 and over , Health Status , Health Status Indicators
2.
Int J Mol Sci ; 25(10)2024 May 13.
Article in English | MEDLINE | ID: mdl-38791345

ABSTRACT

Doxorubicin (DOX) is a potent chemotherapeutic agent known for its multi-organ toxicity, especially in the heart, which limits its clinical application. The toxic side effects of DOX, including DNA damage, oxidative stress, mitochondrial dysfunction and cell apoptosis, are intricately linked to the involvement of nicotinamide adenine dinucleotide (NAD+). To assess the effectiveness of the NAD+ precursor nicotinamide mononucleotide (NMN) in counteracting the multi-organ toxicity of DOX, a mouse model was established through DOX administration, which led to significant reductions in NAD+ in tissues with evident injury, including the heart, liver and lungs. NMN treatment alleviated both multi-organ fibrosis and mortality in mice. Mechanistically, tissue fibrosis, macrophage infiltration and DOX-related cellular damage, which are potentially implicated in the development of multi-organ fibrosis, could be attenuated by NAD+ restoration. Our findings provide compelling evidence for the benefits of NMN supplementation in mitigating the adverse effects of chemotherapeutic drugs on multiple organs.


Subject(s)
Doxorubicin , Fibrosis , Nicotinamide Mononucleotide , Animals , Doxorubicin/adverse effects , Nicotinamide Mononucleotide/pharmacology , Mice , Dietary Supplements , Male , NAD/metabolism , Oxidative Stress/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology
3.
J Adv Res ; 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38479571

ABSTRACT

INTRODUCTION: Chemotherapy-induced peripheral neuropathy (CIPN) is a common complication that affects an increasing number of cancer survivors. However, the current treatment options for CIPN are limited. Paclitaxel (PTX) is a widely used chemotherapeutic drug that induces senescence in cancer cells. While previous studies have demonstrated that Sonic hedgehog (Shh) can counteract cellular dysfunction during aging, its role in CIPN remains unknown. OBJECTIVES: Herein, the aim of this study was to investigate whether Shh activation could inhibits neuronal/glial senescence and alleviates CIPN. METHODS: We treated ND7/23 neuronal cells and RSC96 Schwann cells with two selective Shh activators (purmorphamine [PUR] and smoothened agonist [SAG]) in the presence of PTX. Additionally, we utilized a CIPN mouse model induced by PTX injection. To assess cellular senescence, we performed a senescence-associated ß-galactosidase (SA-ß-gal) assay, measured reactive oxygen species (ROS) levels, and examined the expression of P16, P21, and γH2AX. To understand the underlying mechanisms, we conducted ubiquitin assays, LC-MS/MS, H&E staining, and assessed protein expression through Western blotting and immunofluorescence staining. RESULTS: In vitro, we observed that Shh activation significantly alleviated the senescence-related decline in multiple functions included SA-ß-gal activity, expression of P16 and P21, cell viability, and ROS accumulation in DRG sensory neurons and Schwann cells after PTX exposure. Furthermore, our in vivo experiments demonstrated that Shh activation significantly reduced axonal degeneration, demyelination, and improved nerve conduction. Mechanistically, we discovered that PTX reduced the protein level of SP1, which was ubiquitinated by the E3 ligase TRIM25 at the lysine 694 (K694), leading to increased CXCL13 expression, and we found that Shh activation inhibited PTX-induced neuronal/glial senescence and CIPN through the TRIM25-SP1-CXCL13 axis. CONCLUSION: These findings provide evidence for the role of PTX-induced senescence in DRG sensory neurons and Schwann cells, suggesting that Shh could be a potential therapeutic target for CIPN.

4.
Antioxidants (Basel) ; 12(12)2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38136238

ABSTRACT

Ferroptosis is involved in various tissue injuries including neurodegeneration, ischemia-reperfusion injury, and acute liver injury. Ferroptosis inhibitors exhibit promising clinical potential in the treatment of various diseases. As a traditional chemical, silymarin has been widely used in healthcare and clinical applications to treat liver injuries in which ferroptosis is involved. Silibinin is the main active ingredient of silymarin. However, the effect of silibinin on ferroptosis and ferroptosis-related diseases remains unclear. Here, we found that silibinin inhibited death in different kinds of cells caused by ferroptosis inducers including RSL3 and erastin. Moreover, silibinin alleviated lipid peroxidation induced by RSL3 without affecting the labile iron pool. Next, the antioxidant activity of silibinin was demonstrated by the DPPH assay. In vivo, silibinin strikingly relieved tissue injuries and ferroptosis in the liver and kidney of glutathione peroxidase 4 (GPX4) knockout C57 BL/6J mice. Moreover, silibinin effectively rescued renal ischemia-reperfusion, a well-known ferroptosis-related disease. In conclusion, our study revealed that silibinin effectively inhibits cell ferroptosis and ferroptosis-related tissue injuries, implicating silibinin as a potential chemical to treat ferroptosis-related diseases.

5.
Cell Mol Neurobiol ; 43(7): 3767-3781, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37526811

ABSTRACT

PPARγ coactivator-1 alpha (PGC-1α) is an essential transcription factor co-activator that regulates gene transcription and neural regeneration. Schwann cells, which are unique glial cells in peripheral nerves that dedifferentiate after peripheral nerve injury (PNI) and are released from degenerative nerves. Wallerian degeneration is a series of stereotypical events that occurs in response to nerve fibers after PNI. The role of PGC-1α in Schwann cell dedifferentiation and Wallerian degeneration is not yet clear. As Wallerian degeneration plays a crucial role in PNI, we conducted a study to determine whether PGC-1α has an effect on peripheral nerve degeneration after injury. We examined the expression of PGC-1α after sciatic nerve crush or transection using Western blotting and found that PGC-1α expression increased after PNI. Then we utilized ex vivo and in vitro models to investigate the effects of PGC-1α inhibition and activation on Schwann cell dedifferentiation and nerve degeneration. Our findings indicate that PGC-1α negatively regulates Schwann cell dedifferentiation and nerve degeneration. Through the use of RNA-seq, siRNA/plasmid transfection and reversal experiments, we identified that PGC-1α targets inhibit the expression of paraoxonase 1 (PON1) during Schwann cell dedifferentiation in degenerated nerves. In summary, PGC-1α plays a crucial role in preventing Schwann cell dedifferentiation and its activation can reduce peripheral nerve degeneration by targeting PON1. PGC-1α inhibits Schwann cell dedifferentiation and peripheral nerve degeneration. PGC-1α negatively regulates Schwann cell dedifferentiation and peripheral nerve degeneration after injury by targeting PON1.


Subject(s)
Aryldialkylphosphatase , Peripheral Nerve Injuries , Humans , Aryldialkylphosphatase/metabolism , Aryldialkylphosphatase/pharmacology , Cell Dedifferentiation , Wallerian Degeneration/metabolism , Wallerian Degeneration/pathology , Schwann Cells , Sciatic Nerve/pathology , Peripheral Nerve Injuries/pathology , Nerve Regeneration/physiology
6.
Nucleic Acids Res ; 50(7): 3764-3776, 2022 04 22.
Article in English | MEDLINE | ID: mdl-35323972

ABSTRACT

The core catalytic unit of telomerase comprises telomerase reverse transcriptase (TERT) and telomerase RNA (TERC). Unlike TERT, which is predominantly expressed in cancer and stem cells, TERC is ubiquitously expressed in normal somatic cells without telomerase activity. However, the functions of TERC in these telomerase-negative cells remain elusive. Here, we reported positive feedback regulation between TERC and the PI3K-AKT pathway that controlled cell proliferation independent of telomerase activity in human fibroblasts. Mechanistically, we revealed that TERC activated the transcription of target genes from the PI3K-AKT pathway, such as PDPK1, by targeting their promoters. Overexpression of PDPK1 partially rescued the deficiency of AKT activation caused by TERC depletion. Furthermore, we found that FOXO1, a transcription factor negatively regulated by the PI3K-AKT pathway, bound to TERC promoter and suppressed its expression. Intriguingly, TERC-induced activation of the PI3K-AKT pathway also played a critical role in the proliferation of activated CD4+ T cells. Collectively, our findings identify a novel function of TERC that regulates the PI3K-AKT pathway via positive feedback to elevate cell proliferation independent of telomerase activity and provide a potential strategy to promote CD4+ T cells expansion that is responsible for enhancing adaptive immune reactions to defend against pathogens and tumor cells.


Subject(s)
RNA , Telomerase , 3-Phosphoinositide-Dependent Protein Kinases/metabolism , Cell Proliferation/genetics , Feedback, Physiological , Humans , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA/genetics , RNA/metabolism , Telomerase/genetics , Telomerase/metabolism
7.
Cell Death Dis ; 12(7): 706, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34267193

ABSTRACT

Ferroptosis, a newly defined mode of regulated cell death caused by unbalanced lipid redox metabolism, is implicated in various tissue injuries and tumorigenesis. However, the role of ferroptosis in stem cells has not yet been investigated. Glutathione peroxidase 4 (GPX4) is a critical suppressor of lipid peroxidation and ferroptosis. Here, we study the function of GPX4 and ferroptosis in hematopoietic stem and progenitor cells (HSPCs) in mice with Gpx4 deficiency in the hematopoietic system. We find that Gpx4 deletion solely in the hematopoietic system has no significant effect on the number and function of HSPCs in mice. Notably, hematopoietic stem cells (HSCs) and hematopoietic progenitor cells lacking Gpx4 accumulated lipid peroxidation and underwent ferroptosis in vitro. α-Tocopherol, the main component of vitamin E, was shown to rescue the Gpx4-deficient HSPCs from ferroptosis in vitro. When Gpx4 knockout mice were fed a vitamin E-depleted diet, a reduced number of HSPCs and impaired function of HSCs were found. Furthermore, increased levels of lipid peroxidation and cell death indicated that HSPCs undergo ferroptosis. Collectively, we demonstrate that GPX4 and vitamin E cooperatively maintain lipid redox balance and prevent ferroptosis in HSPCs.


Subject(s)
Antioxidants/pharmacology , Ferroptosis/drug effects , Hematopoietic Stem Cells/drug effects , Lipid Peroxidation/drug effects , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Vitamin E Deficiency/drug therapy , Vitamin E/pharmacology , Animals , Cells, Cultured , Disease Models, Animal , Female , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/enzymology , Hematopoietic Stem Cells/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Vitamin E Deficiency/enzymology , Vitamin E Deficiency/genetics , Vitamin E Deficiency/pathology
8.
Cell Death Differ ; 28(3): 1062-1075, 2021 03.
Article in English | MEDLINE | ID: mdl-33082515

ABSTRACT

Telomeres are transcribed into telomeric RNA termed as TERRA. However, the transcription itself and excessive TERRA may interfere with telomere replication during S phase. The mechanism that coordinates telomere transcription and replication is unknown. Here, we report that TCOF1 leaves the nucleolus and is recruited to telomeres specifically during S phase by interacting with TRF2. Therein, TCOF1 acts to suppress telomere transcription by binding and inhibiting Pol II. Thus, TERRA is limited to low levels in S phase. Depletion of TCOF1 leads to abnormally elevated TERRA and formation of DNA/RNA hybrids (R-loops) at telomeres, which induces replication fork stalling and fragile telomeres. Importantly, telomere replication defect induced by TCOF1 deficiency can be rescued by either masking TERRA or expressing an R-loop eraser RNase H1, demonstrating a critical role of TCOF1 in coordinating telomere transcription and replication. These findings link nucleolus to telomeres and uncover a novel function of TCOF1 on ensuring telomere integrity.


Subject(s)
Nuclear Proteins/genetics , Phosphoproteins/genetics , Ribonuclease H/metabolism , Telomere/metabolism , Telomeric Repeat Binding Protein 2/genetics , Cell Line , DNA Damage , DNA Replication , DNA-Binding Proteins/metabolism , Humans , Telomere/chemistry , Transcription Factors/metabolism
9.
Cell Rep ; 32(2): 107877, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32668244

ABSTRACT

Evolutionarily conserved SCAN (named after SRE-ZBP, CTfin51, AW-1, and Number 18 cDNA)-domain-containing zinc finger transcription factors (ZSCAN) have been found in both mouse and human genomes. Zscan4 is transiently expressed during zygotic genome activation (ZGA) in preimplantation embryos and induced pluripotent stem cell (iPSC) reprogramming. However, little is known about the mechanism of Zscan4 underlying these processes of cell fate control. Here, we show that Zscan4f, a representative of ZSCAN proteins, is able to recruit Tet2 through its SCAN domain. The Zscan4f-Tet2 interaction promotes DNA demethylation and regulates the expression of target genes, particularly those encoding glycolytic enzymes and proteasome subunits. Zscan4f regulates metabolic rewiring, enhances proteasome function, and ultimately promotes iPSC generation. These results identify Zscan4f as an important partner of Tet2 in regulating target genes and promoting iPSC generation and suggest a possible and common mechanism shared by SCAN family transcription factors to recruit ten-eleven translocation (TET) DNA dioxygenases to regulate diverse cellular processes, including reprogramming.


Subject(s)
Cellular Reprogramming/genetics , DNA-Binding Proteins/metabolism , Proteostasis/genetics , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Animals , Base Sequence , DNA/metabolism , DNA-Binding Proteins/genetics , Dioxygenases , Glycolysis/genetics , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/metabolism , MCF-7 Cells , Mice, Inbred C57BL , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Domains , Proto-Oncogene Proteins/genetics , Up-Regulation
10.
PLoS Genet ; 16(6): e1008799, 2020 06.
Article in English | MEDLINE | ID: mdl-32502208

ABSTRACT

TRF2 and TRF1 are a key component in shelterin complex that associates with telomeric DNA and protects chromosome ends. BRM is a core ATPase subunit of SWI/SNF chromatin remodeling complex. Whether and how BRM-SWI/SNF complex is engaged in chromatin end protection by telomeres is unknown. Here, we report that depletion of BRM does not affect heterochromatin state of telomeres, but results in telomere dysfunctional phenomena including telomere uncapping and replication defect. Mechanistically, expression of TRF2 and TRF1 is jointly regulated by BRM-SWI/SNF complex, which is localized to promoter region of both genes and facilitates their transcription. BRM-deficient cells bear increased TRF2-free or TRF1-free telomeres due to insufficient expression. Importantly, BRM depletion-induced telomere uncapping or replication defect can be rescued by compensatory expression of exogenous TRF2 or TRF1, respectively. Together, these results identify a new function of BRM-SWI/SNF complex in enabling functional telomeres for maintaining genome stability.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Telomere/metabolism , Telomeric Repeat Binding Protein 1/genetics , Telomeric Repeat Binding Protein 2/genetics , Transcription Factors/metabolism , Genomic Instability , HEK293 Cells , HeLa Cells , Hep G2 Cells , Heterochromatin/metabolism , Humans , Promoter Regions, Genetic , Telomeric Repeat Binding Protein 1/metabolism , Telomeric Repeat Binding Protein 2/metabolism , Transcription Factors/genetics
11.
Protein Cell ; 11(1): 60-70, 2020 01.
Article in English | MEDLINE | ID: mdl-31350723

ABSTRACT

Pluripotent stem cells (PSCs) such as embryonic stem cells (ESCs), ESCs derived by somatic cell nuclear transfer (ntESCs), and induced pluripotent stem cells (iPSCs) have unlimited capacity for self-renewal and pluripotency and can give rise to all types of somatic cells. In order to maintain their self-renewal and pluripotency, PSCs need to preserve their telomere length and homeostasis. In recent years, increasing studies have shown that telomere reprogramming is essential for stem cell pluripotency maintenance and its induced pluripotency process. Telomere-associated proteins are not only required for telomere maintenance in both stem cells, their extra-telomeric functions have also been found to be critical as well. Here, we will discuss how telomeres and telomere-associated factors participate and regulate the maintenance of stem cell pluripotency.


Subject(s)
Cellular Reprogramming , Pluripotent Stem Cells , Telomerase/metabolism , Telomere Homeostasis , Telomere/metabolism , Animals , Histones/metabolism , Humans , Mice , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Telomere-Binding Proteins/metabolism
12.
Nucleic Acids Res ; 47(15): 8084-8095, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31294790

ABSTRACT

TERC is an RNA component of telomerase. However, TERC is also ubiquitously expressed in most human terminally differentiated cells, which don't have telomerase activity. The function of TERC in these cells is largely unknown. Here, we report that TERC enhances the expression and secretion of inflammatory cytokines by stimulating NK-κB pathway in a telomerase-independent manner. The ectopic expression of TERC in telomerase-negative cells alters the expression of 431 genes with high enrichment of those involved in cellular immunity. We perform genome-wide screening using a previously identified 'binding motif' of TERC and identify 14 genes that are transcriptionally regulated by TERC. Among them, four genes (LIN37, TPRG1L, TYROBP and USP16) are demonstrated to stimulate the activation of NK-κB pathway. Mechanistically, TERC associates with the promoter of these genes through forming RNA-DNA triplexes, thereby enhancing their transcription. In vivo, expression levels of TERC and TERC target genes (TYROBP, TPRG1L and USP16) are upregulated in patients with inflammation-related diseases such as type II diabetes and multiple sclerosis. Collectively, these results reveal an unknown function of TERC on stimulating inflammatory response and highlight a new mechanism by which TERC modulates gene transcription. TERC may be a new target for the development of anti-inflammation therapeutics.


Subject(s)
Cytokines/genetics , Inflammation Mediators/metabolism , RNA/genetics , Telomerase/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cell Line, Tumor , Cytokines/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Promoter Regions, Genetic/genetics , Proteins/genetics , Proteins/metabolism , RNA/metabolism , RNA Interference , Signal Transduction/genetics , Telomerase/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism
13.
Protein Cell ; 10(12): 927-928, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31187422

ABSTRACT

In the original publication the labels in figure 4C and D are incorrectly published. The correct labels for figure 4C and D is provided in this correction.

14.
Protein Cell ; 10(11): 808-824, 2019 11.
Article in English | MEDLINE | ID: mdl-31115790

ABSTRACT

While the majority of all human cancers counteract telomere shortening by expressing telomerase, ~15% of all cancers maintain telomere length by a telomerase-independent mechanism known as alternative lengthening of telomeres (ALT). Here, we show that high load of intrinsic DNA damage is present in ALT cancer cells, leading to apoptosis stress by activating p53-independent, but JNK/c-Myc-dependent apoptotic pathway. Notably, ALT cells expressing wild-type p53 show much lower apoptosis than p53-deficient ALT cells. Mechanistically, we find that intrinsic DNA damage in ALT cells induces low level of p53 that is insufficient to initiate the transcription of apoptosis-related genes, but is sufficient to stimulate the expression of key components of mTORC2 (mTOR and Rictor), which in turn leads to phosphorylation of AKT. Activated AKT (p-AKT) thereby stimulates downstream anti-apoptotic events. Therefore, p53 and AKT are the key factors that suppress spontaneous apoptosis in ALT cells. Indeed, inhibition of p53 or AKT selectively induces rapid death of ALT cells in vitro, and p53 inhibitor severely suppresses the growth of ALT-cell xenograft tumors in mice. These findings reveal a previously unrecognized function of p53 in anti-apoptosis and identify that the inhibition of p53 or AKT has a potential as therapeutics for specifically targeting ALT cancers.


Subject(s)
Proto-Oncogene Proteins c-akt/metabolism , Telomere/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Cell Line, Tumor , DNA Damage , Heterografts , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/therapy , Telomere Homeostasis
15.
J Biol Chem ; 294(16): 6645-6656, 2019 04 19.
Article in English | MEDLINE | ID: mdl-30819801

ABSTRACT

Linear chromosome ends are capped by telomeres that have been previously reported to adopt a t-loop structure. The lack of simple methods for detecting t-loops has hindered progress in understanding the dynamics of t-loop formation and its function in protecting chromosome ends. Here, we employed a classical two-dimensional agarose gel method (2D gel method) to innovatively apply to t-loop detection. Briefly, restriction fragments of genomic DNA were separated in a 2D gel, and the telomere sequence was detected by in-gel hybridization with telomeric probe. Using this method, we found that t-loops are present throughout the cell cycle, and t-loop formation tightly couples to telomere replication. We also observed that t-loop abundance positively correlates with chromatin condensation, i.e. cells with less compact telomeric chromatin (ALT cells and trichostatin A (TSA)-treated HeLa cells) exhibited fewer t-loops. Moreover, we observed that telomere dysfunction-induced foci, ALT-associated promyelocytic leukemia bodies, and telomere sister chromatid exchanges are activated upon TSA-induced loss of t-loops. These findings confirm the importance of the t-loop in protecting linear chromosomes from damage or illegitimate recombination.


Subject(s)
Cell Cycle/physiology , Chromatids/metabolism , Heterochromatin/metabolism , Telomere/metabolism , Chromatids/chemistry , Electrophoresis, Gel, Two-Dimensional , HeLa Cells , Heterochromatin/chemistry , Humans , Hydroxamic Acids/pharmacology , Telomere/chemistry
16.
Aging (Albany NY) ; 11(2): 741-755, 2019 01 28.
Article in English | MEDLINE | ID: mdl-30694216

ABSTRACT

Telomere length and telomere shortening rate (TSR) are accepted indicators of aging in cross-sectional population studies. This study aimed to investigate the potential influence of common antidiabetic agents on telomere length and TSR in patients with type 2 diabetes mellitus (T2DM). Leukocyte telomere length was measured through terminal restriction fragment analysis, and TSR was calculated in 388 T2DM patients. Depending on whether or not they received antidiabetic medication, patients were first divided into a treatment group and a nontreatment group. Treated patients were further subdivided into an acarbose-free group (patients taking antidiabetic agents without acarbose) and an acarbose group (patients using acarbose for more than 3 months). Results showed that untreated patients had higher TSRs than patients on antidiabetic drugs. Interestingly, patients in the acarbose group had significantly higher TSRs than patients in the acarbose-free group. Compared to the nontreatment group, the acarbose group showed better glycemic control of HbA1c, but the TSR was also higher. Our results suggest that antidiabetic treatments without acarbose can slow aging. By contrast, acarbose may accelerate biological aging in patients with T2DM, independently of glycemic control.


Subject(s)
Acarbose/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/therapeutic use , Leukocytes/drug effects , Telomere Shortening , Adult , Cross-Sectional Studies , Female , Humans , Male , Middle Aged
17.
Cell Death Dis ; 9(6): 662, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29855463

ABSTRACT

Blood cells are challenged by intrinsic and exogenous stress that may result in many types of damage to DNA. As a major participant in cell-mediated immunity in blood, T lymphocytes are maintained in their quiescent (resting) state for most of their lives and switch to the proliferating state once stimulated. How resting and stimulated T cells address DNA damage remains largely unknown. Here, we report that while different types of DNA damage are efficiently repaired in stimulated T cells, they result in massive apoptosis of resting T cells. Mechanistically, DNA damage in resting T cells activates the ATM/ATR/DNA-PKcs signaling pathway but fails to induce the formation of γH2AX and 53BP1 foci, leading to unrepaired DNA damage that activates apoptosis in a p53-independent but JNK/p73-dependent manner. Mice challenged with high DNA damage stress display far fewer T cells in peripheral blood, lymph nodes, and spleens. Collectively, these results reveal that resting T cells are hypersensitive to DNA damage due to defects in DNA damage repair mechanisms. These findings provide new insight into T-cell function and maintenance of immunity under highly stressed conditions.


Subject(s)
DNA Damage , DNA Repair , T-Lymphocytes/pathology , Animals , Apoptosis/drug effects , Bleomycin/pharmacology , Cell Line , DNA Breaks, Double-Stranded/drug effects , DNA Repair/drug effects , Female , Fibroblasts/drug effects , Fibroblasts/pathology , Humans , Mice, Inbred BALB C , Mice, Inbred C57BL , Signal Transduction , T-Lymphocytes/drug effects , Tumor Suppressor Protein p53/metabolism
18.
Mol Cell ; 70(3): 395-407.e4, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29727616

ABSTRACT

Telomeres and telomere-binding proteins form complex secondary nucleoprotein structures that are critical for genome integrity but can present serious challenges during telomere DNA replication. It remains unclear how telomere replication stress is resolved during S phase. Here, we show that the BUB3-BUB1 complex, a component in spindle assembly checkpoint, binds to telomeres during S phase and promotes telomere DNA replication. Loss of the BUB3-BUB1 complex results in telomere replication defects, including fragile and shortened telomeres. We also demonstrate that the telomere-binding ability of BUB3 and kinase activity of BUB1 are indispensable to BUB3-BUB1 function at telomeres. TRF2 targets BUB1-BUB3 to telomeres, and BUB1 can directly phosphorylate TRF1 and promote TRF1 recruitment of BLM helicase to overcome replication stress. Our findings have uncovered previously unknown roles for the BUB3-BUB1 complex in S phase and shed light on how proteins from diverse pathways function coordinately to ensure proper telomere replication and maintenance.


Subject(s)
Cell Cycle Proteins/genetics , DNA Replication/genetics , Poly-ADP-Ribose Binding Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Telomere/genetics , Cell Line , Cell Line, Tumor , DNA Helicases/genetics , HEK293 Cells , HeLa Cells , Humans , M Phase Cell Cycle Checkpoints/genetics , S Phase/genetics , Spindle Apparatus/genetics , Telomere-Binding Proteins/genetics
19.
Cell Chem Biol ; 23(10): 1261-1270, 2016 Oct 20.
Article in English | MEDLINE | ID: mdl-27693060

ABSTRACT

G-quadruplexes are specialized secondary structures in nucleic acids that possess significant conformational polymorphisms. The precise G-quadruplex conformations in vivo and their relevance to biological functions remain controversial and unclear, especially for telomeric G-quadruplexes. Here, we report a novel single-chain variable fragment (scFv) antibody, D1, with high binding selectivity for parallel G-quadruplexes in vitro and in vivo. Genome-wide chromatin immunoprecipitation using D1 and deep-sequencing revealed the consensus sequence for parallel G-quadruplex formation, which is characterized by G-rich sequence with a short loop size (<3 nt). By using D1, telomeric parallel G-quadruplex was identified and its formation was regulated by small molecular ligands targeting and telomere replication. Together, parallel G-quadruplex specific antibody D1 was found to be a valuable tool for determination of G-quadruplex and its conformation, which will prompt further studies on the structure of G-quadruplex and its biological implication in vivo.


Subject(s)
G-Quadruplexes , Single-Chain Antibodies/chemistry , Telomere/chemistry , Base Sequence , Cell Line , Consensus Sequence , Genome, Human , HeLa Cells , Humans , Ligands , Models, Molecular , Single-Chain Antibodies/immunology , Telomere/genetics , Telomere/immunology
20.
Nucleic Acids Res ; 44(17): 8086-96, 2016 09 30.
Article in English | MEDLINE | ID: mdl-27220467

ABSTRACT

The majority of tumor cells overcome proliferative limit by expressing telomerase. Whether or not telomerase preferentially extends the shortest telomeres is still under debate. When human cancer cells are cultured at neutral pH, telomerase extends telomeres in telomere length-independent manner. However, the microenvironment of tumor is slightly acidic, and it is not yet known how this influences telomerase action. Here, we examine telomere length homeostasis in tumor cells cultured at pHe 6.8. The results indicate that telomerase preferentially extends short telomeres, such that telomere length distribution narrows and telomeres become nearly uniform in size. After growth at pHe 6.8, the expression of telomerase, TRF1, TRF2 and TIN2 decreases, and the abundance of Cajal bodies decreases. Therefore, telomerase are insufficient for extending every telomere and shorter telomeres bearing less shelterin proteins are more accessible for telomerase recruitment. The findings support the 'protein-counting mechanism' in which extended and unextended state of telomere is determined by the number of associated shelterin proteins and the abundance of telomerase. Decreased expression of telomerase and preferential extension of short telomeres have important implications for tumor cell viability, and generate a strong rationale for research on telomerase-targeted anti-cancer therapeutics.


Subject(s)
Extracellular Space/metabolism , Telomere Shortening , Telomere/metabolism , Acetylation , Chromatin/metabolism , DNA/metabolism , HeLa Cells , Histones/metabolism , Humans , Hydrogen-Ion Concentration , Shelterin Complex , Telomerase/metabolism , Telomere-Binding Proteins/metabolism , Telomeric Repeat Binding Protein 2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...