Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Transplant Cell Ther ; 29(11): 704.e1-704.e8, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37625594

RESUMO

Higher body mass index (BMI) is characterized as a chronic inflammatory state with endothelial dysfunction. Endothelial injury after allogeneic hematopoietic stem cell transplantation (allo-HSCT) puts patients at risk for such complications as transplantation-associated thrombotic microangiopathy (TA-TMA) and acute graft-versus-host-disease (aGVHD). To evaluate the impact of increased BMI on endothelial injury after allo-HSCT in pediatric and young adult patients, we conducted a retrospective cohort study evaluating 476 consecutive allo-HSCT children and young adult recipients age 0 to 20 years. Our analysis was subdivided based on distinct age categories (<2 years and 2 to 20 years). BMI was considered as a variable but was also expressed in standard deviations from the mean adjusted for age and sex (z-score), based on established criteria from the World Health Organization (age <2 years) and the Centers for Disease Control and Prevention (age 2 to 20 years) to account for differences associated with age. Primary endpoints included the incidences of TA-TMA and aGVHD. Increased BMI z-score was associated with TA-TMA after allo-HSCT in patients age <2 years (median, 18.1; IQR, 17 to 20; P = .006) and in patients age 2 to 20 years (median, 18.7; IQR, 16 to 21.9; P = .02). Higher BMI z-score correlated with TA-TMA risk in both age groups, with a BMI z-score of .9 in the younger cohort and .7 (IQR, -.4 to 1.6; P = .04) in the older cohort. Increased BMI z-score was associated with an increased risk of TA-TMA in a multivariate analysis of the entire cohort (odds ratio [OR], 1.2; 95% confidence interval [CI], 1.05 to 1.37; P = .008). Multivariate analysis also demonstrated that patients with BMI in the 85th percentile or greater had an increased risk of developing TA-TMA compared to those with a lower BMI percentile (OR, 2.66; 95% CI, 1.62 to 4.32; P < .001). Baseline and day +7 ST2 levels were elevated in subjects with TA-TMA compared to those without TA-TMA in both age groups. Baseline sC5b-9 concentration was not correlated with BMI z-score, but sC5b-9 concentration was increased markedly by 7 days post-allo-HSCT in patients age <2 years who later developed TA-TMA compared to those who never developed TA-TMA (P = .001). The median BMI z-score was higher for patients with aGVHD compared to patients without aGVHD (.7 [range, -3.9 to 3.9] versus .2 [range, -7.8 to 5.4]; P = .03). We show that high BMI is associated with augmented risk of endothelial injury after HSCT, specifically TA-TMA. These data identify a high-risk population likely to benefit from early interventions to prevent endothelial injury and prompt treatment of established endothelial injury.


Assuntos
Doença Enxerto-Hospedeiro , Transplante de Células-Tronco Hematopoéticas , Microangiopatias Trombóticas , Estados Unidos , Adulto Jovem , Humanos , Criança , Recém-Nascido , Lactente , Pré-Escolar , Adolescente , Adulto , Estudos Retrospectivos , Índice de Massa Corporal , Microangiopatias Trombóticas/complicações , Fatores de Risco , Transplante de Células-Tronco Hematopoéticas/efeitos adversos
2.
Blood Adv ; 7(17): 5137-5151, 2023 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-37083597

RESUMO

Intestinal permeability may correlate with adverse outcomes during hematopoietic stem cell transplantation (HSCT), but longitudinal quantification with traditional oral mannitol and lactulose is not feasible in HSCT recipients because of mucositis and diarrhea. A modified lactulose:rhamnose (LR) assay is validated in children with environmental enteritis. Our study objective was to quantify peri-HSCT intestinal permeability changes using the modified LR assay. The LR assay was administered before transplant, at day +7 and +30 to 80 pediatric and young adult patients who received allogeneic HSCT. Lactulose and rhamnose were detected using urine mass spectrometry and expressed as an L:R ratio. Metagenomic shotgun sequencing of stool for microbiome analyses and enzyme-linked immunosorbent assay analyses of plasma lipopolysaccharide binding protein (LBP), ST2, REG3α, claudin1, occludin, and intestinal alkaline phosphatase were performed at the same timepoints. L:R ratios were increased at day +7 but returned to baseline at day +30 in most patients (P = .014). Conditioning regimen intensity did not affect the trajectory of L:R (P = .39). Baseline L:R ratios did not vary with diagnosis. L:R correlated with LBP levels (r2 = 0.208; P = .0014). High L:R ratios were associated with lower microbiome diversity (P = .035), loss of anaerobic organisms (P = .020), and higher plasma LBP (P = .0014). No adverse gastrointestinal effects occurred because of LR. Intestinal permeability as measured through L:R ratios after allogeneic HSCT correlates with intestinal dysbiosis and elevated plasma LBP. The LR assay is well-tolerated and may identify transplant recipients who are more likely to experience adverse outcomes.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Lactulose , Adulto Jovem , Humanos , Criança , Lactulose/metabolismo , Ramnose , Reação de Fase Aguda , Disbiose , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Permeabilidade
3.
Stem Cell Reports ; 17(4): 734-740, 2022 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-35276089

RESUMO

The intestine is one of the organs that relies on stem cell function for maintaining tissue homeostasis. Recent findings on intestinal aging show that intestinal architecture, such as villus length, crypt size, and cell composition changes in the aged crypts. The correspondent decline in the regenerative capacity of the intestine is mainly due to a decline in intestinal stem cell function upon aging, as the underlying mechanisms of aging intestinal stem cells are beginning to unravel. This review summarizes our current knowledge on stem cell-intrinsic mechanisms of aging of intestinal stem cells and their connection to extrinsic factors, such as niche cells and microbiota and will introduce recent approaches to attenuate or even revert the aging of intestinal stem cells.


Assuntos
Intestinos , Células-Tronco , Homeostase , Mucosa Intestinal
4.
iScience ; 24(4): 102362, 2021 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-33870147

RESUMO

Homeostasis in the intestinal epithelium is maintained by Lgr5-positive intestinal stem cells (ISCs) located at the base of the crypt. The function of ISCs is reduced upon aging which leads to a decline of regeneration of the intestinal epithelium. We report that aged intestinal crypts present with an elevated activity of the small RhoGTPase Cdc42. Elevation of Cdc42 activity in young animals by genetic means causes premature ISC aging, whereas pharmacological suppression of elevated Cdc42 activity restores organoid formation potential in vitro. Consistent with a critical role of elevated Cdc42 activity in aged ISCs for a reduced regenerative capacity of aged ISCs, suppression of Cdc42 activity in vivo improves crypt regeneration in aged mice. Thus, pharmacological reduction of Cdc42 activity can improve the regeneration of aged intestinal epithelium.

5.
Aging (Albany NY) ; 13(4): 4778-4793, 2021 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-33629967

RESUMO

Normal hair growth occurs in cycles, comprising growth (anagen), cessation (catagen) and rest (telogen). Upon aging, the initiation of anagen is significantly delayed, which results in impaired hair regeneration. Hair regeneration is driven by hair follicle stem cells (HFSCs). We show here that aged HFSCs present with a decrease in canonical Wnt signaling and a shift towards non-canonical Wnt5a driven signaling which antagonizes canonical Wnt signaling. Elevated expression of Wnt5a in HFSCs upon aging results in elevated activity of the small RhoGTPase Cdc42 as well as a change in the spatial distribution of Cdc42 within HFSCs. Treatment of aged HFSC with a specific pharmacological inhibitor of Cdc42 activity termed CASIN to suppress the aging-associated elevated activity of Cdc42 restored canonical Wnt signaling in aged HFSCs. Treatment of aged mice in vivo with CASIN induced anagen onset and increased the percentage of anagen skin areas. Aging-associated functional deficits of HFSCs are at least in part intrinsic to HFSCs and can be restored by rational pharmacological approaches.


Assuntos
Folículo Piloso/crescimento & desenvolvimento , Rejuvenescimento/fisiologia , Células-Tronco/metabolismo , Via de Sinalização Wnt , Proteína Wnt-5a/genética , Animais , Senescência Celular/fisiologia , Camundongos
7.
Methods Mol Biol ; 2171: 41-52, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32705634

RESUMO

Aging is a multifactorial process. Organ maintenance and tissue regeneration are impaired upon aging mainly due to loss of stem cell function in organs that depend on stem cell in the adult. Intestine is such an organ, and upon aging intestinal regeneration is impaired due to decline of intestinal stem cell function. To determine the aging status of intestine and intestinal stem cells, histological analyses; analyses of the level of proliferation markers in tissue by immunofluorescence and/or quantitative RT-PCR; and gene expression analysis for stemness related genes in isolated crypts, intestinal stem cells (ISC), and Paneth cells can be used. To analyze the level of regeneration in intestine and thus determine a decline in ISC function, techniques like in vitro organoid cultures and lineage tracing with BrdU, lineage tracing using transgenic mice and histological analyses of tissue regeneration after 3 and 5 days after two rounds of 10 Gy of radiation (a 10 + 10 Gy IR experiment) can be applied. In this chapter we will focus on protocols for lineage tracing, the 10 + 10 gy IR experiment and for organoid cultures from young and aged mouse intestine.Lineage tracing experiments in intestine can be done in many ways. In this chapter we describe a protocol for lineage tracing upon BrdU incorporation and lineage tracing using the Lgr5eGFPCreERT2 Rosa26YFP transgenic mouse. For BrdU based-lineage tracing BrdU is administrated via intraperitoneal injections into mice. Animals will be analyzed 3 days (72 h) after BrdU administration. For experiments involving Lgr5eGFPCreERT2 Rosa26YFP mice, mice will be analyzed after tamoxifen injection that activates Cre in Lgr5 positive (ISC) cells, which will result in permanent YFP expression. This allows for tracing of YFP positive cells in the intestine. The time point for the analysis of the intestinal tissue will depend in this case on the underlying scientific question that will be addressed. For 10 + 10 Gy experiments, animals will be irradiated with a radiation dose of 10 Gy on 2 consecutive days. The intestinal tissue will be analyzed 3 and 5 days after the second dose of radiation. Quantitative analyses of crypt depth and determination of the rate of crypt fission upon histochemistry will provide an estimation on the in vivo regenerative potential of ISCs. For serial organoid culture experiments, crypts will be harvested from mouse intestine, initially plated at concentrations ranging from 500 to 1000 crypts per well in Matrigel and grown in conditional medium or ISC medium. ISC Medium is changed every 2 days. After 1 week in culture, the organoids will be disrupted via a syringe and replated in fresh Matrigel. As the ability to form multilobed organoids is considered to be a direct stem cell function, the frequency of organoid formation in serial replating experiments can serve as a quantitative measurement of ISC function. For example, we demonstrated a reduced frequency of organoid formation as well as a reduction in number of lobes formed per organoid after 4-5 replatings of intestinal organoids from aged compared to young mice. These three techniques are thus, in combination, able to quantify the regeneration potential of intestinal stem cells and thus determine the extent to which intestinal stem cell regenerative function is reduced upon aging.


Assuntos
Intestinos/citologia , Células-Tronco/citologia , Animais , Células Cultivadas , Feminino , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Masculino , Camundongos , Organoides/citologia , Organoides/metabolismo
8.
Mol Cell ; 78(1): 141-151.e5, 2020 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-32027840

RESUMO

Polycomb repressive complex 2 (PRC2) places H3K27me3 at developmental genes and is causally implicated in keeping bivalent genes silent. It is unclear if that silence requires minimum H3K27me3 levels and how the mark transmits faithfully across mammalian somatic cell generations. Mouse intestinal cells lacking EZH2 methyltransferase reduce H3K27me3 proportionately at all PRC2 target sites, but ∼40% uniform residual levels keep target genes inactive. These genes, derepressed in PRC2-null villus cells, remain silent in intestinal stem cells (ISCs). Quantitative chromatin immunoprecipitation and computational modeling indicate that because unmodified histones dilute H3K27me3 by 50% each time DNA replicates, PRC2-deficient ISCs initially retain sufficient H3K27me3 to avoid gene derepression. EZH2 mutant human lymphoma cells also require multiple divisions before H3K27me3 dilution relieves gene silencing. In both cell types, promoters with high basal H3K4me2/3 activate in spite of some residual H3K27me3, compared to less-poised promoters. These findings have implications for PRC2 inhibition in cancer therapy.


Assuntos
Replicação do DNA , Proteína Potenciadora do Homólogo 2 de Zeste/fisiologia , Inativação Gênica , Código das Histonas , Regiões Promotoras Genéticas , Animais , Linhagem Celular Tumoral , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Histonas/metabolismo , Humanos , Intestinos/citologia , Camundongos , Complexo Repressor Polycomb 2/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Células-Tronco/metabolismo , Ativação Transcricional
9.
Stem Cell Reports ; 9(6): 1961-1975, 2017 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-29129684

RESUMO

RHOA, a founding member of the Rho GTPase family, is critical for actomyosin dynamics, polarity, and morphogenesis in response to developmental cues, mechanical stress, and inflammation. In murine small intestinal epithelium, inducible RHOA deletion causes a loss of epithelial polarity, with disrupted villi and crypt organization. In the intestinal crypts, RHOA deficiency results in reduced cell proliferation, increased apoptosis, and a loss of intestinal stem cells (ISCs) that mimic effects of radiation damage. Mechanistically, RHOA loss reduces YAP signaling of the Hippo pathway and affects YAP effector epiregulin (EREG) expression in the crypts. Expression of an active YAP (S112A) mutant rescues ISC marker expression, ISC regeneration, and ISC-associated Wnt signaling, but not defective epithelial polarity, in RhoA knockout mice, implicating YAP in RHOA-regulated ISC function. EREG treatment or active ß-catenin Catnblox(ex3) mutant expression rescues the RhoA KO ISC phenotypes. Thus, RHOA controls YAP-EREG signaling to regulate intestinal homeostasis and ISC regeneration.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Epirregulina/genética , Intestino Delgado/metabolismo , Morfogênese/genética , Fosfoproteínas/genética , Proteínas rho de Ligação ao GTP/genética , Animais , Proteínas de Ciclo Celular , Diferenciação Celular/genética , Proliferação de Células/genética , Epirregulina/metabolismo , Epitélio/crescimento & desenvolvimento , Epitélio/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Intestino Delgado/crescimento & desenvolvimento , Camundongos , Camundongos Knockout , Células-Tronco/citologia , Células-Tronco/metabolismo , Via de Sinalização Wnt/genética , Proteínas de Sinalização YAP , beta Catenina/genética , Proteína rhoA de Ligação ao GTP
10.
Cell Rep ; 18(11): 2608-2621, 2017 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-28297666

RESUMO

Although intestinal homeostasis is maintained by intestinal stem cells (ISCs), regeneration is impaired upon aging. Here, we first uncover changes in intestinal architecture, cell number, and cell composition upon aging. Second, we identify a decline in the regenerative capacity of ISCs upon aging because of a decline in canonical Wnt signaling in ISCs. Changes in expression of Wnts are found in stem cells themselves and in their niche, including Paneth cells and mesenchyme. Third, reactivating canonical Wnt signaling enhances the function of both murine and human ISCs and, thus, ameliorates aging-associated phenotypes of ISCs in an organoid assay. Our data demonstrate a role for impaired Wnt signaling in physiological aging of ISCs and further identify potential therapeutic avenues to improve ISC regenerative potential upon aging.


Assuntos
Senescência Celular , Intestino Delgado/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Via de Sinalização Wnt , Animais , Biomarcadores/metabolismo , Contagem de Células , Proliferação de Células , Feminino , Camundongos , Organoides/citologia , Regeneração , Nicho de Células-Tronco
11.
Cell ; 165(6): 1389-1400, 2016 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-27212235

RESUMO

Bivalent promoters in embryonic stem cells (ESCs) carry methylation marks on two lysine residues, K4 and K27, in histone3 (H3). K4me2/3 is generally considered to promote transcription, and Polycomb Repressive Complex 2 (PRC2) places K27me3, which is erased at lineage-restricted genes when ESCs differentiate in culture. Molecular defects in various PRC2 null adult tissues lack a unifying explanation. We found that epigenomes in adult mouse intestine and other self-renewing tissues show fewer and distinct bivalent promoters compared to ESCs. Groups of tissue-specific genes that carry bivalent marks are repressed, despite the presence of promoter H3K4me2/3. These are the predominant genes de-repressed in PRC2-deficient adult cells, where aberrant expression is proportional to the H3K4me2/3 levels observed at their promoters in wild-type cells. Thus, in adult animals, PRC2 specifically represses genes with acquired, tissue-restricted promoter bivalency. These findings provide new insights into specificity in chromatin-based gene regulation.


Assuntos
Células-Tronco Embrionárias/metabolismo , Complexo Repressor Polycomb 2/genética , Regiões Promotoras Genéticas , Animais , Diferenciação Celular/genética , Metilação de DNA , Regulação da Expressão Gênica , Histonas/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/citologia , Lisina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Complexo Repressor Polycomb 2/metabolismo
13.
Nature ; 506(7489): 511-5, 2014 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-24413398

RESUMO

Cells differentiate when transcription factors bind accessible cis-regulatory elements to establish specific gene expression programs. In differentiating embryonic stem cells, chromatin at lineage-restricted genes becomes sequentially accessible, probably by means of 'pioneer' transcription factor activity, but tissues may use other strategies in vivo. Lateral inhibition is a pervasive process in which one cell forces a different identity on its neighbours, and it is unclear how chromatin in equipotent progenitors undergoing lateral inhibition quickly enables distinct, transiently reversible cell fates. Here we report the chromatin and transcriptional underpinnings of differentiation in mouse small intestine crypts, where notch signalling mediates lateral inhibition to assign progenitor cells into absorptive or secretory lineages. Transcript profiles in isolated LGR5(+) intestinal stem cells and secretory and absorptive progenitors indicated that each cell population was distinct and the progenitors specified. Nevertheless, secretory and absorptive progenitors showed comparable levels of H3K4me2 and H3K27ac histone marks and DNase I hypersensitivity--signifying accessible, permissive chromatin-at most of the same cis-elements. Enhancers acting uniquely in progenitors were well demarcated in LGR5(+) intestinal stem cells, revealing early priming of chromatin for divergent transcriptional programs, and retained active marks well after lineages were specified. On this chromatin background, ATOH1, a secretory-specific transcription factor, controls lateral inhibition through delta-like notch ligand genes and also drives the expression of numerous secretory lineage genes. Depletion of ATOH1 from specified secretory cells converted them into functional enterocytes, indicating prolonged responsiveness of marked enhancers to the presence or absence of a key transcription factor. Thus, lateral inhibition and intestinal crypt lineage plasticity involve interaction of a lineage-restricted transcription factor with broadly permissive chromatin established in multipotent stem cells.


Assuntos
Diferenciação Celular/genética , Cromatina/genética , Cromatina/metabolismo , Regulação da Expressão Gênica , Intestino Delgado/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Linhagem da Célula/genética , Desoxirribonuclease I/metabolismo , Elementos Facilitadores Genéticos/genética , Enterócitos/citologia , Enterócitos/metabolismo , Feminino , Histonas/metabolismo , Intestino Delgado/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores Notch/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Transcrição Gênica
14.
Nat Cell Biol ; 14(1): 73-9, 2011 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-22138576

RESUMO

The tumour suppressor p53 activates Puma-dependent apoptosis and p21-dependent cell-cycle arrest in response to DNA damage. Deletion of p21 improved stem-cell function and organ maintenance in progeroid mice with dysfunctional telomeres, but the function of Puma has not been investigated in this context. Here we show that deletion of Puma improves stem- and progenitor-cell function, organ maintenance and lifespan of telomere-dysfunctional mice. Puma deletion impairs the clearance of stem and progenitor cells that have accumulated DNA damage as a consequence of critically short telomeres. However, further accumulation of DNA damage in these rescued progenitor cells leads to increasing activation of p21. RNA interference experiments show that upregulation of p21 limits proliferation and evolution of chromosomal imbalances of Puma-deficient stem and progenitor cells with dysfunctional telomeres. These results provide experimental evidence that p53-dependent apoptosis and cell-cycle arrest act in cooperating checkpoints limiting tissue maintenance and evolution of chromosomal instability at stem- and progenitor-cell levels in response to telomere dysfunction. Selective inhibition of Puma-dependent apoptosis can result in temporary improvements in maintenance of telomere-dysfunctional organs.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Pontos de Checagem do Ciclo Celular/genética , Instabilidade Cromossômica , Inibidor de Quinase Dependente de Ciclina p21/genética , Células-Tronco/fisiologia , Telômero/genética , Proteínas Supressoras de Tumor/genética , Animais , Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Processos de Crescimento Celular/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Dano ao DNA , Camundongos , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Células-Tronco/metabolismo , Telômero/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Regulação para Cima
15.
PLoS One ; 6(2): e17122, 2011 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-21364961

RESUMO

INTRODUCTION: Telomere shortening is a cell-intrinsic mechanism that limits cell proliferation by induction of DNA damage responses resulting either in apoptosis or cellular senescence. Shortening of telomeres has been shown to occur during human aging and in chronic diseases that accelerate cell turnover, such as chronic hepatitis. Telomere shortening can limit organ homeostasis and regeneration in response to injury. Whether the same holds true for pancreas regeneration in response to injury is not known. METHODS: In the present study, pancreatic regeneration after acute cerulein-induced pancreatitis was studied in late generation telomerase knockout mice with short telomeres compared to telomerase wild-type mice with long telomeres. RESULTS: Late generation telomerase knockout mice exhibited impaired exocrine pancreatic regeneration after acute pancreatitis as seen by persistence of metaplastic acinar cells and markedly reduced proliferation. The expression levels of p53 and p21 were not significantly increased in regenerating pancreas of late generation telomerase knockout mice compared to wild-type mice. CONCLUSION: Our results indicate that pancreatic regeneration is limited in the context of telomere dysfunction without evidence for p53 checkpoint activation.


Assuntos
Pâncreas Exócrino/fisiologia , Regeneração/genética , Telomerase/genética , Telômero/metabolismo , Animais , Divisão Celular/genética , Senescência Celular/genética , Dano ao DNA/genética , Dano ao DNA/fisiologia , Camundongos , Camundongos Knockout , Pâncreas Exócrino/metabolismo , RNA/genética , Regeneração/fisiologia , Telomerase/metabolismo , Telomerase/fisiologia , Telômero/genética , Telômero/patologia , Telômero/fisiologia , Fatores de Tempo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteína Supressora de Tumor p53/fisiologia
16.
EMBO Rep ; 11(8): 619-25, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20577265

RESUMO

Telomere shortening limits the proliferation of primary human fibroblasts by the induction of senescence, which is mediated by ataxia telangiectasia mutated-dependent activation of p53. Here, we show that CHK2 deletion impairs the induction of senescence in mouse and human fibroblasts. By contrast, CHK2 deletion did not improve the stem-cell function, organ maintenance and lifespan of telomere dysfunctional mice and did not prevent the induction of p53/p21, apoptosis and cell-cycle arrest in telomere dysfunctional progenitor cells. Together, these results indicate that CHK2 mediates the induction of senescence in fibroblasts, but is dispensable for the induction of telomere dysfunction checkpoints at the stem and progenitor cell level in vivo.


Assuntos
Proteínas Serina-Treonina Quinases/metabolismo , Células-Tronco/fisiologia , Telômero/metabolismo , Animais , Ciclo Celular/fisiologia , Senescência Celular/fisiologia , Quinase do Ponto de Checagem 2 , Dano ao DNA , Fibroblastos/citologia , Fibroblastos/fisiologia , Humanos , Mucosa Intestinal/metabolismo , Intestinos/citologia , Camundongos , Camundongos Knockout , Proteínas Serina-Treonina Quinases/genética , Células-Tronco/citologia , Telomerase/genética , Telomerase/metabolismo
17.
Nat Genet ; 41(10): 1138-43, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19718028

RESUMO

Telomere dysfunction limits the proliferative capacity of human cells and induces organismal aging by activation of p53 and p21. Although deletion of p21 elongates the lifespan of telomere-dysfunctional mice, a direct analysis of p53 in telomere-related aging has been hampered by early tumor formation in p53 knockout mice. Here we analyzed the functional consequences of conditional p53 deletion. Intestinal deletion of p53 shortened the lifespan of telomere-dysfunctional mice without inducing tumor formation. In contrast to p21 deletion, the deletion of p53 impaired the depletion of chromosomal-instable intestinal stem cells in aging telomere-dysfunctional mice. These instable stem cells contributed to epithelial regeneration leading to an accumulation of chromosomal instability, increased apoptosis, altered epithelial cell differentiation and premature intestinal failure. Together, these results provide the first experimental evidence for an organ system in which p53-dependent mechanisms prevent tissue destruction in response to telomere dysfunction by depleting genetically instable stem cells.


Assuntos
Envelhecimento/fisiologia , Instabilidade Cromossômica , Deleção de Genes , Células-Tronco/metabolismo , Telômero/genética , Proteína Supressora de Tumor p53/deficiência , Animais , Ciclo Celular , Dano ao DNA , Genoma , Mucosa Intestinal/metabolismo , Intestinos/citologia , Camundongos , Camundongos Knockout , Células-Tronco/citologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
18.
Exp Gerontol ; 43(11): 998-1004, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18817864

RESUMO

The decline in organ maintenance and function is one of the major problems limiting quality of life during aging. The accumulation of telomere dysfunction and DNA damage appears to be one of the underlying causes. Uncapping of chromosome ends in response to critical telomere shortening limits the proliferative capacity of human cells by activation of DNA damage checkpoints inducing senescence or apoptosis. Telomere shortening occurs in the vast majority of human tissues during aging and in chronic diseases that increase the rate of cell turnover. There is emerging evidence that telomere shortening can limit the maintenance and function of adult stem cells -- a cell type of utmost importance for organ maintenance and regeneration. In mouse models, telomere dysfunction leads to a depletion of adult stem cell compartments suggesting that stem cells are very sensitive to DNA damage. Both the rarity of stem and progenitor cells in adult organs and their removal in response to damage make it difficult to assess the impact of telomere dysfunction and DNA damage on stem and progenitor cell aging. Such approaches require the development of sensitive biomarkers recognizing low levels of telomere dysfunction and DNA damage in stem and progenitor cells. Here, we review experimental data on the prevalence of telomere dysfunction and DNA damage during aging and its possible impact on stem and progenitor cell aging.


Assuntos
Células-Tronco Adultas/citologia , Senescência Celular/fisiologia , Células-Tronco/citologia , Telômero/metabolismo , Células-Tronco Adultas/enzimologia , Animais , Peptídeos Catiônicos Antimicrobianos/análise , Biomarcadores/análise , Catelicidinas , Quitinases/análise , Dano ao DNA , Humanos , Camundongos , Camundongos Knockout , Modelos Animais , Fator 1 de Elongação de Peptídeos/análise , Estatmina/análise , Células-Tronco/enzimologia , Telomerase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...