Cookies help us deliver our services. By using our services, you agree to our use of cookies. More information

Difference between revisions of "Guachalla 2009 Aging (Albany NY)"

From Bioblast
 
(19 intermediate revisions by 8 users not shown)
Line 1: Line 1:
{{Publication
{{Publication
|title=Guachalla LM, Ju Z, Koziel R, Figura G, Song Z, Fusser M, Epe B,Jansen-Dürr P,Rudolph KL (2009) Sod2 haploinsufficiency does not accelerate aging of telomere dysfunctional mice. Aging 1: 3.
|title=Guachalla LM, Ju Z, Koziel R, Figura G, Song Z, Fusser M, Epe B, Jansen-Dürr P, Rudolph KL (2009) SOD2 haploinsufficiency does not accelerate aging of telomere dysfunctional mice. Aging (Albany NY) 1:303-15.
|authors=Guachalla LM, Ju Z, Koziel R, Figura G, Song Z, Fusser M, Epe B,Jansen-Duerr P,Rudolph KL
|info=[http://www.ncbi.nlm.nih.gov/pubmed/20195488 PMID: 20195488 Open Access]
|authors=Guachalla Luis Miguel, Ju Zhenyu, Koziel Rafal, Figura Guido von, Song Zhangfa, Fusser Markus, Epe Bernd, Jansen-Duerr Pidder, Rudolph K Lenhard
|year=2009
|year=2009
|journal=Aging
|journal=Aging (Albany NY)
|abstract=Telomere shortening represents a causal factor of cellular senescence. At the same time, several lines of evidenceindicate a pivotal role of oxidative DNA damage for the aging process ''in vivo''. A causal connection between the twoobservations was suggested by experiments showing accelerated telomere shorting under conditions of oxidative stress incultured cells, but has never been studied ''in vivo''. We therefore have analysed whether an increase in mitochondrialderived oxidative stress in response to heterozygous deletion of superoxide dismutase (''Sod2+/‐'') would exacerbate agingphenotypes in telomere dysfunctional (''mTerc‐/‐'') mice. Heterozygous deletion of ''Sod2'' resulted in reduced SOD2 proteinlevels and increased oxidative stress in aging telomere dysfunctional mice, but this did not lead to an increase in basallevels of oxidative nuclear DNA damage, an accumulation of nuclear DNA breaks, or an increased rate of telomereshortening in the mice. Moreover, heterozygous deletion of ''Sod2'' did not accelerate the depletion of stem cells and theimpairment in organ maintenance in aging ''mTerc‐/‐'' mice. In agreement with these observations, ''Sod2'' haploinsufficiencydid not lead to a further reduction in lifespan of mTerc‐/‐ mice. Together, these results indicate that a decrease in SOD2‐dependent antioxidant defence does not exacerbate aging in the context of telomere dysfunction.
|abstract=Telomere shortening represents a causal factor of cellular senescence. At the same time, several lines of evidenceindicate a pivotal role of oxidative DNA damage for the aging process ''in vivo''. A causal connection between the two observations was suggested by experiments showing accelerated telomere shorting under conditions of oxidative stress in cultured cells, but has never been studied ''in vivo''. We therefore have analysed whether an increase in mitochondrial-derived oxidative stress in response to heterozygous deletion of superoxide dismutase (''Sod2+/‐'') would exacerbate aging phenotypes in telomere dysfunctional (''mTerc‐/‐'') mice. Heterozygous deletion of ''Sod2'' resulted in reduced SOD2 protein levels and increased oxidative stress in aging telomere dysfunctional mice, but this did not lead to an increase in basal levels of oxidative nuclear DNA damage, an accumulation of nuclear DNA breaks, or an increased rate of telomere shortening in the mice. Moreover, heterozygous deletion of ''Sod2'' did not accelerate the depletion of stem cells and the impairment in organ maintenance in aging ''mTerc‐/‐'' mice. In agreement with these observations, ''Sod2'' haploinsufficiency did not lead to a further reduction in lifespan of mTerc‐/‐ mice. Together, these results indicate that a decrease in SOD2‐dependent antioxidant defence does not exacerbate aging in the context of telomere dysfunction.
|keywords=Oxidative stress, Superoxide, Telomere shortening, Aging, DNA damage, SOD2, Free radicals, Stem cells
|keywords=Oxidative stress, Superoxide, Telomere shortening, Aging, DNA damage, SOD2, Free radicals, Stem cells
|info=[http://www.ncbi.nlm.nih.gov/pubmed/20195488 PMID: 20195488]
|mipnetlab=AT Innsbruck Jansen-Duerr P
}}
}}
{{Labeling
{{Labeling
|injuries=Aging; Senescence, Genetic Defect; Knockdown; Overexpression
|area=Respiration, Genetic knockout;overexpression
|organism=Mouse
|organism=Mouse
|tissues=Neurons; Brain
|tissues=Nervous system
|preparations=Intact Cell; Cultured; Primary
|preparations=Intact cells
|enzymes=Uncoupler Protein
|enzymes=Uncoupling protein
|kinetics=Inhibitor; Uncoupler
|diseases=Aging;senescence
|topics=Respiration; OXPHOS; ETS Capacity, Redox State
|topics=Inhibitor, Redox state
|couplingstates=ROUTINE
|instruments=Oxygraph-2k
|instruments=Oxygraph-2k
|articletype=Protocol; Manual
}}
}}

Latest revision as of 09:00, 8 March 2020

Publications in the MiPMap
Guachalla LM, Ju Z, Koziel R, Figura G, Song Z, Fusser M, Epe B, Jansen-Dürr P, Rudolph KL (2009) SOD2 haploinsufficiency does not accelerate aging of telomere dysfunctional mice. Aging (Albany NY) 1:303-15.

» PMID: 20195488 Open Access

Guachalla Luis Miguel, Ju Zhenyu, Koziel Rafal, Figura Guido von, Song Zhangfa, Fusser Markus, Epe Bernd, Jansen-Duerr Pidder, Rudolph K Lenhard (2009) Aging (Albany NY)

Abstract: Telomere shortening represents a causal factor of cellular senescence. At the same time, several lines of evidenceindicate a pivotal role of oxidative DNA damage for the aging process in vivo. A causal connection between the two observations was suggested by experiments showing accelerated telomere shorting under conditions of oxidative stress in cultured cells, but has never been studied in vivo. We therefore have analysed whether an increase in mitochondrial-derived oxidative stress in response to heterozygous deletion of superoxide dismutase (Sod2+/‐) would exacerbate aging phenotypes in telomere dysfunctional (mTerc‐/‐) mice. Heterozygous deletion of Sod2 resulted in reduced SOD2 protein levels and increased oxidative stress in aging telomere dysfunctional mice, but this did not lead to an increase in basal levels of oxidative nuclear DNA damage, an accumulation of nuclear DNA breaks, or an increased rate of telomere shortening in the mice. Moreover, heterozygous deletion of Sod2 did not accelerate the depletion of stem cells and the impairment in organ maintenance in aging mTerc‐/‐ mice. In agreement with these observations, Sod2 haploinsufficiency did not lead to a further reduction in lifespan of mTerc‐/‐ mice. Together, these results indicate that a decrease in SOD2‐dependent antioxidant defence does not exacerbate aging in the context of telomere dysfunction. Keywords: Oxidative stress, Superoxide, Telomere shortening, Aging, DNA damage, SOD2, Free radicals, Stem cells

O2k-Network Lab: AT Innsbruck Jansen-Duerr P


Labels: MiParea: Respiration, Genetic knockout;overexpression  Pathology: Aging;senescence 

Organism: Mouse  Tissue;cell: Nervous system  Preparation: Intact cells  Enzyme: Uncoupling protein  Regulation: Inhibitor, Redox state  Coupling state: ROUTINE 

HRR: Oxygraph-2k