Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Aging (Albany NY) ; 15(11): 4625-4641, 2023 06 07.
Article in English | MEDLINE | ID: mdl-37294548

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the coronavirus disease 2019 (COVID-19) pandemic. The severity of COVID-19 increases with each decade of life, a phenomenon that suggest that organismal aging contributes to the fatality of the disease. In this regard, we and others have previously shown that COVID-19 severity correlates with shorter telomeres, a molecular determinant of aging, in patient's leukocytes. Lung injury is a predominant feature of acute SARS-CoV-2 infection that can further progress to lung fibrosis in post-COVID-19 patients. Short or dysfunctional telomeres in Alveolar type II (ATII) cells are sufficient to induce pulmonary fibrosis in mouse and humans. Here, we analyze telomere length and the histopathology of lung biopsies from a cohort of alive post-COVID-19 patients and a cohort of age-matched controls with lung cancer. We found loss of ATII cellularity and shorter telomeres in ATII cells concomitant with a marked increase in fibrotic lung parenchyma remodeling in post- COVID-19 patients compared to controls. These findings reveal a link between presence of short telomeres in ATII cells and long-term lung fibrosis sequel in Post-COVID-19 patients.


Subject(s)
COVID-19 , Neoplasms , Pulmonary Fibrosis , Humans , Mice , Animals , Pulmonary Fibrosis/pathology , COVID-19/pathology , SARS-CoV-2 , Alveolar Epithelial Cells , Lung/pathology , Neoplasms/pathology , Telomere/pathology
2.
Cell Death Differ ; 30(6): 1585-1600, 2023 06.
Article in English | MEDLINE | ID: mdl-37085672

ABSTRACT

Non-small cell lung cancer (NSCLC) is a leading cause of cancer death. Tumor progression depends on interactions of cancer cells with the tumor microenvironment. Here, we find increased copy number and mRNA expression of the catalytic subunit of telomerase, TERT, in tumors from NSCLC patients, contributing to a lower survival. Moreover, TERT expression in NSCLC patients from the TCGA cohort is mainly associated to the reduced infiltration of CD8+ T lymphocytes, as well as to increased infiltration of myeloid-derived suppressor cells (MDSCs). We also show that TERT deficiency and dysfunctional telomeres induced by 6-thio-dG treatment in mice reduced lung tumor implantation and vascularization, increased DNA damage response, cell cycle arrest and apoptosis, as well as reduced proliferation, inflammation, lung tumor immunosupression and invasion upon induction of a Lewis lung carcinoma (LLC). Furthermore, 6-thio-dG-treated human NSCLC xenografts exhibited increased telomere damage, cell cycle arrest and apoptosis, as well as reduced proliferation, resulting in a reduced tumor growth. Our results show that targeting telomeres might be an effective therapeutic strategy in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Telomerase , Humans , Mice , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Telomerase/genetics , Telomerase/metabolism , Heterografts , Tumor Microenvironment , Telomere/metabolism , Lung/metabolism , Cell Line, Tumor
3.
Cell Cycle ; 22(23-24): 2505-2521, 2023.
Article in English | MEDLINE | ID: mdl-38219218

ABSTRACT

Pluripotent stem cells (PSCs) are a promising source of stem cells for regenerative therapies. Stem cell function depends on telomere maintenance mechanisms that provide them with the proliferative capacity and genome stability necessary to multiply and regenerate tissues. We show here that established human embryonic stem cells (hESCs) have stable telomere length that is dependent on telomerase but not on alternative mechanisms based on homologous recombination pathways. Here, we show that human-induced pluripotent stem cells (hiPSCs) reprogrammed from somatic cells show progressive telomere lengthening until reaching a length similar to ESCs. hiPSCs also acquire telomeric chromatin marks of ESCs including decreased abundance of tri-methylated histone H3K9 and H4K20 and HP1 heterochromatic marks, as well as of the shelterin component TRF2. These chromatin features are accompanied with increased abundance of telomere transcripts or TERRAs. We also found that telomeres of both hESCs and hiPSCs are well protected from DNA damage during telomere elongation and once full telomere length is achieved, and exhibit stable genomes. Collectively, this study highlights that hiPSCs acquire ESC features during reprogramming and reveals the telomere biology in human pluripotent stem cells (hPSCs).


We show that established human embryonic stem cells (hESCs) have a maximum and stable telomere length that is dependent on telomerase but not on the alternative homologous recombination pathway or ALT. Human-induced pluripotent stem cells (hiPSCs) reprogrammed from somatic cells show progressive telomere lengthening until reaching a length similar maximum telomere length than ESCs, suggesting that telomere length is regulated by epigenetic mechanisms in human cells. In this regard, hiPSCs acquire telomeric chromatin marks characteristic of an "open chromatin" including increased abundance of telomere transcripts or TERRAs. Telomeres of both hESCs and hiPSCs are well protected during telomere elongation and exhibit stable genomes. Collectively, this study highlights that hiPSCs acquire ESC features during reprogramming and reveals the telomere biology in human pluripotent stem cells (hiPSCs).


Subject(s)
Pluripotent Stem Cells , Telomerase , Humans , Pluripotent Stem Cells/metabolism , Telomere Homeostasis , Histones/metabolism , Telomere/metabolism , Telomerase/genetics , Telomerase/metabolism , Chromatin/metabolism
4.
PLoS Genet ; 18(6): e1010260, 2022 06.
Article in English | MEDLINE | ID: mdl-35727838

ABSTRACT

The shelterin protein POT1 has been found mutated in many different familial and sporadic cancers, however, no mouse models to understand the pathobiology of these mutations have been developed so far. To address the molecular mechanisms underlying the tumorigenic effects of POT1 mutant proteins in humans, we have generated a mouse model for the human POT1R117C mutation found in Li-Fraumeni-Like families with cases of cardiac angiosarcoma by introducing this mutation in the Pot1a endogenous locus, knock-in for Pot1aR117C. We find here that both mouse embryonic fibroblasts (MEFs) and tissues from Pot1a+/ki mice show longer telomeres than wild-type controls. Longer telomeres in Pot1a+/ki MEFs are dependent on telomerase activity as they are not found in double mutant Pot1a+/ki Tert-/- telomerase-deficient MEFs. By using complementation assays we further show that POT1a pR117C exerts dominant-negative effects at telomeres. As in human Li-Fraumeni patients, heterozygous Pot1a+/ki mice spontaneously develop a high incidence of angiosarcomas, including cardiac angiosarcomas, and this is associated to the presence of abnormally long telomeres in endothelial cells as well as in the tumors. The Pot1a+/R117C mouse model constitutes a useful tool to understand human cancers initiated by POT1 mutations.


Subject(s)
Hemangiosarcoma , Shelterin Complex/metabolism , Telomerase , Telomere-Binding Proteins/metabolism , Animals , Endothelial Cells/metabolism , Fibroblasts/metabolism , Hemangiosarcoma/genetics , Humans , Li-Fraumeni Syndrome , Mice , Mutation , Telomerase/genetics , Telomerase/metabolism , Telomere/genetics , Telomere/metabolism , Telomere-Binding Proteins/genetics
5.
PLoS Genet ; 17(3): e1009410, 2021 03.
Article in English | MEDLINE | ID: mdl-33690611

ABSTRACT

The telomere-bound shelterin complex is essential for chromosome-end protection and genomic stability. Little is known on the regulation of shelterin components by extracellular signals including developmental and environmental cues. Here, we show that human TRF1 is subjected to AKT-dependent regulation. To study the importance of this modification in vivo, we generate knock-in human cell lines carrying non-phosphorylatable mutants of the AKT-dependent TRF1 phosphorylation sites by CRISPR-Cas9. We find that TRF1 mutant cells show decreased TRF1 binding to telomeres and increased global and telomeric DNA damage. Human cells carrying non-phosphorylatable mutant TRF1 alleles show accelerated telomere shortening, demonstrating that AKT-dependent TRF1 phosphorylation regulates telomere maintenance in vivo. TRF1 mutant cells show an impaired response to proliferative extracellular signals as well as a decreased tumorigenesis potential. These findings indicate that telomere protection and telomere length can be regulated by extracellular signals upstream of PI3K/AKT activation, such as growth factors, nutrients or immune regulators, and this has an impact on tumorigenesis potential.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Telomere/genetics , Telomere/metabolism , Animals , DNA Damage , Genomic Instability , Humans , Mice , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Telomere Shortening , Telomeric Repeat Binding Protein 1/genetics , Telomeric Repeat Binding Protein 1/metabolism
6.
Aging (Albany NY) ; 13(1): 1-15, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33428591

ABSTRACT

The incidence of severe manifestations of COVID-19 increases with age with older patients showing the highest mortality, suggesting that molecular pathways underlying aging contribute to the severity of COVID-19. One mechanism of aging is the progressive shortening of telomeres, which are protective structures at chromosome ends. Critically short telomeres impair the regenerative capacity of tissues and trigger loss of tissue homeostasis and disease. The SARS-CoV-2 virus infects many different cell types, forcing cell turn-over and regeneration to maintain tissue homeostasis. We hypothesize that presence of short telomeres in older patients limits the tissue response to SARS-CoV-2 infection. We measure telomere length in peripheral blood lymphocytes COVID-19 patients with ages between 29 and 85 years-old. We find that shorter telomeres are associated to increased severity of the disease. Individuals within the lower percentiles of telomere length and higher percentiles of short telomeres have higher risk of developing severe COVID-19 pathologies.


Subject(s)
Aging/genetics , COVID-19/genetics , Telomere Shortening , Telomere/genetics , Adult , Age Factors , Aged , Aged, 80 and over , COVID-19/diagnosis , Female , Humans , Male , Middle Aged , Risk Assessment , Risk Factors , Severity of Illness Index , COVID-19 Drug Treatment
SELECTION OF CITATIONS
SEARCH DETAIL
...