Bone fragility and decline in stem cells in prematurely aging DNA repair deficient trichothiodystrophy mice.

Standard

Bone fragility and decline in stem cells in prematurely aging DNA repair deficient trichothiodystrophy mice. / Diderich, Karin E M; Nicolaije, Claudia; Priemel, Matthias; Waarsing, Jan H; Day, Judd S; Brandt, Renata M C; Schilling, Arndt; Botter, Sander M; Weinans, Harrie; Horst, van der; Gijsbertus, T J; Hoeijmakers, Jan H J; Leeuwen, van; Johannes, P T M.

in: AGE (DORDR), Jahrgang 34, Nr. 4, 4, 2012, S. 845-861.

Publikationen: SCORING: Beitrag in Fachzeitschrift/ZeitungSCORING: ZeitschriftenaufsatzForschungBegutachtung

Harvard

Diderich, KEM, Nicolaije, C, Priemel, M, Waarsing, JH, Day, JS, Brandt, RMC, Schilling, A, Botter, SM, Weinans, H, Horst, VD, Gijsbertus, TJ, Hoeijmakers, JHJ, Leeuwen, V & Johannes, PTM 2012, 'Bone fragility and decline in stem cells in prematurely aging DNA repair deficient trichothiodystrophy mice.', AGE (DORDR), Jg. 34, Nr. 4, 4, S. 845-861. <http://www.ncbi.nlm.nih.gov/pubmed/21814739?dopt=Citation>

APA

Diderich, K. E. M., Nicolaije, C., Priemel, M., Waarsing, J. H., Day, J. S., Brandt, R. M. C., Schilling, A., Botter, S. M., Weinans, H., Horst, V. D., Gijsbertus, T. J., Hoeijmakers, J. H. J., Leeuwen, V., & Johannes, P. T. M. (2012). Bone fragility and decline in stem cells in prematurely aging DNA repair deficient trichothiodystrophy mice. AGE (DORDR), 34(4), 845-861. [4]. http://www.ncbi.nlm.nih.gov/pubmed/21814739?dopt=Citation

Vancouver

Diderich KEM, Nicolaije C, Priemel M, Waarsing JH, Day JS, Brandt RMC et al. Bone fragility and decline in stem cells in prematurely aging DNA repair deficient trichothiodystrophy mice. AGE (DORDR). 2012;34(4):845-861. 4.

Bibtex

@article{cef0ee9def1c4854b668fd3d1f840fc4,
title = "Bone fragility and decline in stem cells in prematurely aging DNA repair deficient trichothiodystrophy mice.",
abstract = "Trichothiodystrophy (TTD) is a rare, autosomal recessive nucleotide excision repair (NER) disorder caused by mutations in components of the dual functional NER/basal transcription factor TFIIH. TTD mice, carrying a patient-based point mutation in the Xpd gene, strikingly resemble many features of the human syndrome and exhibit signs of premature aging. To examine to which extent TTD mice resemble the normal process of aging, we thoroughly investigated the bone phenotype. Here, we show that female TTD mice exhibit accelerated bone aging from 39 weeks onwards as well as lack of periosteal apposition leading to reduced bone strength. Before 39 weeks have passed, bones of wild-type and TTD mice are identical excluding a developmental defect. Albeit that bone formation is decreased, osteoblasts in TTD mice retain bone-forming capacity as in vivo PTH treatment leads to increased cortical thickness. In vitro bone marrow cell cultures showed that TTD osteoprogenitors retain the capacity to differentiate into osteoblasts. However, after 13 weeks of age TTD females show decreased bone nodule formation. No increase in bone resorption or the number of osteoclasts was detected. In conclusion, TTD mice show premature bone aging, which is preceded by a decrease in mesenchymal stem cells/osteoprogenitors and a change in systemic factors, identifying DNA damage and repair as key determinants for bone fragility by influencing osteogenesis and bone metabolism.",
keywords = "Animals, Female, Age Factors, Sensitivity and Specificity, Immunohistochemistry, Disease Models, Animal, Mice, Mice, Inbred C57BL, Microscopy, Electron, Analysis of Variance, Reference Values, Random Allocation, Osteogenesis/drug effects/genetics, Osteoblasts/metabolism, Osteoclasts/metabolism, Aging, Premature/*genetics/physiopathology, Bone and Bones/drug effects/*pathology/ultrastructure, DNA Damage/drug effects, DNA Repair-Deficiency Disorders/*genetics, Hematopoietic Stem Cells/*metabolism, Osteoporosis/genetics/physiopathology, Parathyroid Hormone/*pharmacology, Trichothiodystrophy Syndromes/*genetics, Animals, Female, Age Factors, Sensitivity and Specificity, Immunohistochemistry, Disease Models, Animal, Mice, Mice, Inbred C57BL, Microscopy, Electron, Analysis of Variance, Reference Values, Random Allocation, Osteogenesis/drug effects/genetics, Osteoblasts/metabolism, Osteoclasts/metabolism, Aging, Premature/*genetics/physiopathology, Bone and Bones/drug effects/*pathology/ultrastructure, DNA Damage/drug effects, DNA Repair-Deficiency Disorders/*genetics, Hematopoietic Stem Cells/*metabolism, Osteoporosis/genetics/physiopathology, Parathyroid Hormone/*pharmacology, Trichothiodystrophy Syndromes/*genetics",
author = "Diderich, {Karin E M} and Claudia Nicolaije and Matthias Priemel and Waarsing, {Jan H} and Day, {Judd S} and Brandt, {Renata M C} and Arndt Schilling and Botter, {Sander M} and Harrie Weinans and Horst, {van der} and Gijsbertus, {T J} and Hoeijmakers, {Jan H J} and van Leeuwen and Johannes, {P T M}",
year = "2012",
language = "English",
volume = "34",
pages = "845--861",
journal = "AGE (DORDR)",
issn = "0161-9152",
publisher = "Springer Netherlands",
number = "4",

}

RIS

TY - JOUR

T1 - Bone fragility and decline in stem cells in prematurely aging DNA repair deficient trichothiodystrophy mice.

AU - Diderich, Karin E M

AU - Nicolaije, Claudia

AU - Priemel, Matthias

AU - Waarsing, Jan H

AU - Day, Judd S

AU - Brandt, Renata M C

AU - Schilling, Arndt

AU - Botter, Sander M

AU - Weinans, Harrie

AU - Horst, van der

AU - Gijsbertus, T J

AU - Hoeijmakers, Jan H J

AU - Leeuwen, van

AU - Johannes, P T M

PY - 2012

Y1 - 2012

N2 - Trichothiodystrophy (TTD) is a rare, autosomal recessive nucleotide excision repair (NER) disorder caused by mutations in components of the dual functional NER/basal transcription factor TFIIH. TTD mice, carrying a patient-based point mutation in the Xpd gene, strikingly resemble many features of the human syndrome and exhibit signs of premature aging. To examine to which extent TTD mice resemble the normal process of aging, we thoroughly investigated the bone phenotype. Here, we show that female TTD mice exhibit accelerated bone aging from 39 weeks onwards as well as lack of periosteal apposition leading to reduced bone strength. Before 39 weeks have passed, bones of wild-type and TTD mice are identical excluding a developmental defect. Albeit that bone formation is decreased, osteoblasts in TTD mice retain bone-forming capacity as in vivo PTH treatment leads to increased cortical thickness. In vitro bone marrow cell cultures showed that TTD osteoprogenitors retain the capacity to differentiate into osteoblasts. However, after 13 weeks of age TTD females show decreased bone nodule formation. No increase in bone resorption or the number of osteoclasts was detected. In conclusion, TTD mice show premature bone aging, which is preceded by a decrease in mesenchymal stem cells/osteoprogenitors and a change in systemic factors, identifying DNA damage and repair as key determinants for bone fragility by influencing osteogenesis and bone metabolism.

AB - Trichothiodystrophy (TTD) is a rare, autosomal recessive nucleotide excision repair (NER) disorder caused by mutations in components of the dual functional NER/basal transcription factor TFIIH. TTD mice, carrying a patient-based point mutation in the Xpd gene, strikingly resemble many features of the human syndrome and exhibit signs of premature aging. To examine to which extent TTD mice resemble the normal process of aging, we thoroughly investigated the bone phenotype. Here, we show that female TTD mice exhibit accelerated bone aging from 39 weeks onwards as well as lack of periosteal apposition leading to reduced bone strength. Before 39 weeks have passed, bones of wild-type and TTD mice are identical excluding a developmental defect. Albeit that bone formation is decreased, osteoblasts in TTD mice retain bone-forming capacity as in vivo PTH treatment leads to increased cortical thickness. In vitro bone marrow cell cultures showed that TTD osteoprogenitors retain the capacity to differentiate into osteoblasts. However, after 13 weeks of age TTD females show decreased bone nodule formation. No increase in bone resorption or the number of osteoclasts was detected. In conclusion, TTD mice show premature bone aging, which is preceded by a decrease in mesenchymal stem cells/osteoprogenitors and a change in systemic factors, identifying DNA damage and repair as key determinants for bone fragility by influencing osteogenesis and bone metabolism.

KW - Animals

KW - Female

KW - Age Factors

KW - Sensitivity and Specificity

KW - Immunohistochemistry

KW - Disease Models, Animal

KW - Mice

KW - Mice, Inbred C57BL

KW - Microscopy, Electron

KW - Analysis of Variance

KW - Reference Values

KW - Random Allocation

KW - Osteogenesis/drug effects/genetics

KW - Osteoblasts/metabolism

KW - Osteoclasts/metabolism

KW - Aging, Premature/genetics/physiopathology

KW - Bone and Bones/drug effects/pathology/ultrastructure

KW - DNA Damage/drug effects

KW - DNA Repair-Deficiency Disorders/genetics

KW - Hematopoietic Stem Cells/metabolism

KW - Osteoporosis/genetics/physiopathology

KW - Parathyroid Hormone/pharmacology

KW - Trichothiodystrophy Syndromes/genetics

KW - Animals

KW - Female

KW - Age Factors

KW - Sensitivity and Specificity

KW - Immunohistochemistry

KW - Disease Models, Animal

KW - Mice

KW - Mice, Inbred C57BL

KW - Microscopy, Electron

KW - Analysis of Variance

KW - Reference Values

KW - Random Allocation

KW - Osteogenesis/drug effects/genetics

KW - Osteoblasts/metabolism

KW - Osteoclasts/metabolism

KW - Aging, Premature/genetics/physiopathology

KW - Bone and Bones/drug effects/pathology/ultrastructure

KW - DNA Damage/drug effects

KW - DNA Repair-Deficiency Disorders/genetics

KW - Hematopoietic Stem Cells/metabolism

KW - Osteoporosis/genetics/physiopathology

KW - Parathyroid Hormone/pharmacology

KW - Trichothiodystrophy Syndromes/genetics

M3 - SCORING: Journal article

VL - 34

SP - 845

EP - 861

JO - AGE (DORDR)

JF - AGE (DORDR)

SN - 0161-9152

IS - 4

M1 - 4

ER -