Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
2.
Nat Commun ; 14(1): 6109, 2023 09 30.
Article in English | MEDLINE | ID: mdl-37777550

ABSTRACT

The influence of aging on intestinal stem cells and their niche can explain underlying causes for perturbation in their function observed during aging. Molecular mechanisms for such a decrease in the functionality of intestinal stem cells during aging remain largely undetermined. Using transcriptome-wide approaches, our study demonstrates that aging intestinal stem cells strongly upregulate antigen presenting pathway genes and over-express secretory lineage marker genes resulting in lineage skewed differentiation into the secretory lineage and strong upregulation of MHC class II antigens in the aged intestinal epithelium. Mechanistically, we identified an increase in proinflammatory cells in the lamina propria as the main source of elevated interferon gamma (IFNγ) in the aged intestine, that leads to the induction of Stat1 activity in intestinal stem cells thus priming the aberrant differentiation and elevated antigen presentation in epithelial cells. Of note, systemic inhibition of IFNγ-signaling completely reverses these aging phenotypes and reinstalls regenerative capacity of the aged intestinal epithelium.


Subject(s)
Interferon-gamma , Intestines , Homeostasis , Interferon-gamma/metabolism , Intestinal Mucosa , Intestines/metabolism , Animals , Mice , STAT1 Transcription Factor/metabolism
3.
BMC Biol ; 21(1): 6, 2023 01 11.
Article in English | MEDLINE | ID: mdl-36627630

ABSTRACT

BACKGROUND: Methods for the long-term in situ transduction of the unperturbed murine intestinal epithelium have not been developed in past research. Such a method could speed up functional studies and screens to identify genetic factors influencing intestinal epithelium biology. Here, we developed an efficient method achieving this long-sought goal. RESULTS: We used ultrasound-guided microinjections to transduce the embryonic endoderm at day 8 (E8.0) in utero. The injection procedure can be completed in 20 min and had a 100% survival rate. By injecting a small volume (0.1-0.2 µl) of concentrated virus, single shRNA constructs as well as lentiviral libraries can successfully be transduced. The new method stably and reproducibly targets adult intestinal epithelium, as well as other endoderm-derived organs such as the lungs, pancreas, liver, stomach, and bladder. Postnatal analysis of young adult mice indicates that single transduced cells at E8.0 gave rise to crypt fields that were comprised of 20-30 neighbouring crypts per crypt-field at 90 days after birth. Lentiviral targeting of ApcMin/+ mutant and wildtype mice revealed that heterozygous loss of Apc function suppresses the developmental normal growth pattern of intestinal crypt fields. This suppression of crypt field sizes did not involve a reduction of the crypt number per field, indicating that heterozygous Apc loss impaired the growth of individual crypts within the fields. Lentiviral-mediated shRNA knockdown of p53 led to an approximately 20% increase of individual crypts per field in both Apc+/+ and ApcMin/+ mice, associating with an increase in crypt size in ApcMin/+ mice but a slight reduction in crypt size in Apc+/+ mice. Overall, p53 knockdown rescued the reduction in crypt field size in Apc-mutant mice but had no effect on crypt field size in wildtype mice. CONCLUSIONS: This study develops a novel technique enabling robust and reproducible in vivo targeting of intestinal stem cells in situ in the unperturbed intestinal epithelium across different regions of the intestine. In vivo somatic gene editing and genetic screening of lentiviral libraries has the potential to speed up discoveries and mechanistic understanding of genetic pathways controlling the biology of the intestinal epithelium during development and postnatal life. The here developed method enables such approaches.


Subject(s)
Genes, APC , Tumor Suppressor Protein p53 , Mice , Animals , Mice, Transgenic , Tumor Suppressor Protein p53/metabolism , Intestinal Mucosa/metabolism , Stem Cells/metabolism
4.
Int J Mol Sci ; 22(18)2021 Sep 10.
Article in English | MEDLINE | ID: mdl-34575966

ABSTRACT

Meiotic defects derived from incorrect DNA repair during gametogenesis can lead to mutations, aneuploidies and infertility. The coordinated resolution of meiotic recombination intermediates is required for crossover formation, ultimately necessary for the accurate completion of both rounds of chromosome segregation. Numerous master kinases orchestrate the correct assembly and activity of the repair machinery. Although much less is known, the reversal of phosphorylation events in meiosis must also be key to coordinate the timing and functionality of repair enzymes. Cdc14 is a crucial phosphatase required for the dephosphorylation of multiple CDK1 targets in many eukaryotes. Mutations that inactivate this phosphatase lead to meiotic failure, but until now it was unknown if Cdc14 plays a direct role in meiotic recombination. Here, we show that the elimination of Cdc14 leads to severe defects in the processing and resolution of recombination intermediates, causing a drastic depletion in crossovers when other repair pathways are compromised. We also show that Cdc14 is required for the correct activity and localization of the Holliday Junction resolvase Yen1/GEN1. We reveal that Cdc14 regulates Yen1 activity from meiosis I onwards, and this function is essential for crossover resolution in the absence of other repair pathways. We also demonstrate that Cdc14 and Yen1 are required to safeguard sister chromatid segregation during the second meiotic division, a late action that is independent of the earlier role in crossover formation. Thus, this work uncovers previously undescribed functions of the evolutionary conserved Cdc14 phosphatase in the regulation of meiotic recombination.


Subject(s)
CDC2 Protein Kinase/genetics , Cell Cycle Proteins/genetics , Holliday Junction Resolvases/genetics , Meiosis/genetics , Protein Tyrosine Phosphatases/genetics , Saccharomyces cerevisiae Proteins/genetics , Chromosome Segregation/genetics , Crossing Over, Genetic/genetics , DNA Repair/genetics , DNA, Cruciform/genetics , Gametogenesis/genetics , Homologous Recombination/genetics , Mutation/genetics , Phosphorylation/genetics , Saccharomyces cerevisiae/genetics
5.
Cell Rep ; 31(4): 107565, 2020 04 28.
Article in English | MEDLINE | ID: mdl-32348758

ABSTRACT

The small intestine is responsible for nutrient absorption and one of the most important interfaces between the environment and the body. During aging, changes of the epithelium lead to food malabsorption and reduced barrier function, thus increasing disease risk. The drivers of these alterations remain poorly understood. Here, we compare the proteomes of intestinal crypts from mice across different anatomical regions and ages. We find that aging alters epithelial immunity, metabolism, and cell proliferation and is accompanied by region-dependent skewing in the cellular composition of the epithelium. Of note, short-term dietary restriction followed by refeeding partially restores the epithelium by promoting stem cell differentiation toward the secretory lineage. We identify Hmgcs2 (3-hydroxy-3-methylglutaryl-coenzyme A [CoA] synthetase 2), the rate-limiting enzyme for ketogenesis, as a modulator of stem cell differentiation that responds to dietary changes, and we provide an atlas of region- and age-dependent proteome changes of the small intestine.


Subject(s)
Diet Therapy/methods , Proteomics/methods , Age Factors , Animals , Humans , Intestinal Mucosa/metabolism , Mice
6.
Gut Microbes ; 12(1): 1714401, 2020 11 09.
Article in English | MEDLINE | ID: mdl-31983316

ABSTRACT

Methotrexate (MTX) is a typical chemotherapeutic drug that is widely used in the treatment of various malignant diseases as well as autoimmune diseases, with gastrointestinal toxicity being its most prominent complication which could have a significant effect on the prognosis of patients. Yet effective ways to alleviate such complications remains to be explored. Here we show that 30% dietary restriction (DR) for 2 weeks dramatically increased the survival rate of 2-month-old female mice after lethal-dose MTX exposure. DR significantly reduced intestinal inflammation, preserved the number of basal crypt PCNA-positive cells, and protected the function of intestinal stem cells (ISCs) after MTX treatment. Furthermore, ablating intestinal microbiota by broad-spectrum antibiotics completely eliminated the protective effect achieved by DR. 16S rRNA gene deep-sequencing analysis revealed that short-term DR significantly increased the Lactobacillus genus, with Lactobacillus rhamnosus GG gavage partially mimicking the rescue effect of DR on the intestines of ad libitum fed mice exposed to lethal-dose MTX. Together, the current study reveals that DR could be a highly effective way to alleviate the lethal injury in the intestine after high-dose MTX treatment, which is functionally mediated by increasing the protective intestinal microbiota taxa in mice. Keywords: Dietary restriction, Methotrexate, Gut microbiota, Intestinal stem cells, intestinal toxicity.


Subject(s)
Antimetabolites, Antineoplastic/toxicity , Caloric Restriction/methods , Gastrointestinal Microbiome/drug effects , Intestinal Mucosa/pathology , Methotrexate/toxicity , Probiotics/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Female , Gastrointestinal Microbiome/physiology , Humans , Inflammation/prevention & control , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology , Lacticaseibacillus rhamnosus/metabolism , Mice , Mice, Inbred C57BL , RNA, Ribosomal, 16S/genetics
7.
Stem Cell Rev Rep ; 16(1): 212-221, 2020 02.
Article in English | MEDLINE | ID: mdl-31797147

ABSTRACT

Aging is characterized by the accumulation of DNA damage and a decrease in stem cell functionality, yet molecular mechanisms that limit the maintenance of stem cells in response to DNA damage remain to be delineated. Here we show in mouse models that DNA damage leads to a transient over-activation of Wnt signaling in hematopoietic stem cells (HSCs), and that high activity of canonical Wnt/ß-catenin signaling sensitizes HSCs to DNA damage induced by X-irradiation which results in preferential maintenance of HSCs with low levels of Wnt signaling. The study shows that genetic or chemical activation of canonical Wnt signaling enhances radiosensitivity of HSCs while inhibition of Wnt signaling decreases it. Together, these results indicate that levels of Wnt signaling activity mediate heterogeneity in the sensitivity of HSCs to DNA damage induced depletion. These findings could be relevant for molecular alterations and selection of stem cells in the context of DNA damage accumulation during aging and cancer formation.


Subject(s)
Hematopoietic Stem Cells/metabolism , Neoplasms/genetics , Stem Cells/metabolism , beta Catenin/genetics , Aging/genetics , Aging/metabolism , Animals , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , DNA Damage/drug effects , DNA Damage/genetics , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/radiation effects , Humans , Mice , Neoplasms/pathology , Radiation Tolerance/genetics , Stem Cells/drug effects , Stem Cells/radiation effects , Wnt Proteins/genetics , Wnt Signaling Pathway/genetics , X-Rays
8.
Biogerontology ; 20(6): 849-855, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31501966

ABSTRACT

In the original publication of the article, the alphabets in figures were published in upper case and mismatched with the figure legends. The corrected figures and figure legends are given in this Correction.

9.
Biogerontology ; 20(6): 837-848, 2019 12.
Article in English | MEDLINE | ID: mdl-31401701

ABSTRACT

The world's aging population is growing rapidly. Incidences of multiple pathologies, such as abdominal obesity, cardiovascular and cerebrovascular diseases, type 2 diabetes, and malignant neoplasms, increase sharply with age. Aged individuals possess a significantly shifted composition of gut microbiota, which is suggested to play important roles in aging associated pathologies. Whether the existing shifted structural composition of microbiota in aged populations can be reverted non-pharmacologically has not been studied so far. Here, we show an intestinal flora imbalance in old C57BL/6J mice with a remarkable dominant proportion of microbes promoting lipid metabolism and inflammation. Intriguingly, short-term (2 months) dietary restriction was enough to significantly revert the imbalance of intestinal flora in aged mice toward a more balanced structural composition as shown in young mice. Our study provides the first evidence that short-term dietary restriction in old mice can restore the already dysfunctional aged gut microbiota. Our study provides the first evidence that short-term dietary restriction in old mice can restore the already dysfunctional aged gut microbiota, which may help ameliorate aging-related disorders plaguing the vast elderly population.


Subject(s)
Aging/metabolism , Energy Intake , Gastrointestinal Microbiome , Animals , Mice , Mice, Inbred C57BL
10.
Stem Cell Rev Rep ; 15(3): 448-455, 2019 06.
Article in English | MEDLINE | ID: mdl-30790135

ABSTRACT

Stem cell aging underlies aging-associated disorders, such as steeply increased incidences of tumors and impaired regeneration capacity upon stress. However, whether and how the intestinal stem cells age remains largely unknown. Here we show that intestinal stem cells derived from 24-month-old mice hardly form typical organoids with crypt-villus structures, but rather mainly form big, rounded cysts devoid of differentiated cell types, which mimics the culturing of heterozygous APC-deficient cells from the APCmin mouse line. Further analysis showed that cultured crypts derived from aged mice exhibited reduced expression levels of differentiation genes and higher expression of Wnt target genes. Lowering the concentration of R-spondin-1 in the culture system significantly reduced formation of rounded cysts, accompanied by an increased formation of organoids from crypts derived from old mice. We are the first to uncover that intestinal stem cells derived from old mice harbor significant deficiency in differentiation that can be partially rescued through a reduction in R-spondin-1 exposure. This could be highly relevant to intestinal tumor development and the reduced regeneration potential observed in the aged population. Our study provides the first experimental evidence that an over-responsiveness to Wnt/beta-catenin signaling of aged intestinal stem cells mediates the aging-induced deficiency in differentiation, and could serve as a potential target to ameliorate aging-associated intestinal pathologies.


Subject(s)
Aging/metabolism , Cell Differentiation , Intestinal Mucosa/metabolism , Stem Cells/metabolism , Wnt Signaling Pathway , Adenomatous Polyposis Coli Protein/deficiency , Adenomatous Polyposis Coli Protein/metabolism , Aging/pathology , Animals , Intestinal Mucosa/pathology , Intestinal Neoplasms/genetics , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Mice , Mice, Mutant Strains , Stem Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...