Lorenz Studer, Elsa Vera, Daniela Cornacchia  Cell Stem Cell 

Slides:



Advertisements
Similar presentations
IPSC Crowdsourcing: A Model for Obtaining Large Panels of Stem Cell Lines for Screening Mahendra Rao Cell Stem Cell Volume 13, Issue 4, Pages (October.
Advertisements

Myc Represses Primitive Endoderm Differentiation in Pluripotent Stem Cells Keriayn N. Smith, Amar M. Singh, Stephen Dalton Cell Stem Cell Volume 7, Issue.
Ann Intern Med. 2011;155(2): doi: / Figure Legend:
Stems Cells and the Pathways to Aging and Cancer
Stem Cells: A Renaissance in Human Biology Research
Volume 14, Issue 6, Pages (June 2014)
Blood Development: Hematopoietic Stem Cell Dependence and Independence
Kevin Andrew Uy Gonzales, Huck-Hui Ng  Cell Stem Cell 
How to Remake a Fibroblast into a Neural Stem Cell
Finding Shangri-La: Limiting the Impact of Senescence on Aging
Valeria V. Orlova, Christine L. Mummery  Cell Stem Cell 
Human Induced Pluripotent Stem Cells: Now Open to Discovery
Cell Cycle Rules Pluripotency
Lotte Bruens, Hugo J.G. Snippert  Cell Stem Cell 
K. Lenhard Rudolph, Daniel Hartmann, Oliver G. Opitz  Gastroenterology 
Reprogramming toward Heart Regeneration: Stem Cells and Beyond
The 3D Genome Shapes Up For Pluripotency
Upgrading from iMac to iMicro
Pluripotent Stem Cells from Cloned Human Embryos: Success at Long Last
Roger A. Barker, Malin Parmar, Lorenz Studer, Jun Takahashi 
Anselme Perrier, Marc Peschanski  Cell Stem Cell 
Pluripotent stem cell models of cardiac disease and their implication for drug discovery and development  Richard P. Davis, Cathelijne W. van den Berg,
Volume 21, Issue 6, Pages (December 2017)
Robert Passier, Christine Mummery  Cell Stem Cell 
Justin Brumbaugh, Konrad Hochedlinger  Cell Stem Cell 
Unveiling the Role of Senescence-Induced Cellular Plasticity
Robert Zweigerdt, Ina Gruh, Ulrich Martin  Cell Stem Cell 
Human Induced Pluripotent Stem Cells: Now Open to Discovery
Psychiatric Disorders: Diagnosis to Therapy
Nat. Rev. Cardiol. doi: /nrcardio
MicroRNA Regulation of Stem Cell Fate
Soledad Matus, Danilo B. Medinas, Claudio Hetz  Cell Stem Cell 
Remodeling Neurodegeneration: Somatic Cell Reprogramming-Based Models of Adult Neurological Disorders  Liang Qiang, Ryousuke Fujita, Asa Abeliovich  Neuron 
Krishanu Saha, Rudolf Jaenisch  Cell Stem Cell 
Adrian Kee Keong Teo, Amy J. Wagers, Rohit N. Kulkarni  Cell Metabolism 
Nat. Rev. Nephrol. doi: /nrneph
Florian T. Merkle, Kevin Eggan  Cell Stem Cell 
Induced Neuronal Cells: How to Make and Define a Neuron
Senescence in Health and Disease
Loukia Yiangou, Alexander D.B. Ross, Kim Jee Goh, Ludovic Vallier 
Direct Lineage Reprogramming: Strategies, Mechanisms, and Applications
Pluripotent Stem Cells and Disease Modeling
Roger A. Barker, Malin Parmar, Lorenz Studer, Jun Takahashi 
Bursts of Reprogramming: A Path to Extend Lifespan?
Immune Modulation of Stem Cells and Regeneration
Order from Chaos: Single Cell Reprogramming in Two Phases
Engineering Stem Cell Organoids
Engineering Hematopoietic Stem Cells: Lessons from Development
Induction of Pluripotency: From Mouse to Human
Psychiatric Disorders: Diagnosis to Therapy
Dynamic Pluripotent Stem Cell States and Their Applications
Steve S.W. Han, Luis A. Williams, Kevin C. Eggan  Neuron 
Blood Development: Hematopoietic Stem Cell Dependence and Independence
Volume 21, Issue 1, Pages (July 2017)
The Stem Cell Niche in Regenerative Medicine
Modeling Rett Syndrome with Stem Cells
Stem Cells: A Renaissance in Human Biology Research
Mitochondrial Regulation in Pluripotent Stem Cells
Direct Conversion Provides Old Neurons from Aged Donor’s Skin
Pluripotent stem cell models of cardiac disease and their implication for drug discovery and development  Richard P. Davis, Cathelijne W. van den Berg,
From Skin to Blood: A New Member Joins the iClub
Cardiac Stem Cell Therapy and the Promise of Heart Regeneration
Abby Sarkar, Konrad Hochedlinger  Cell Stem Cell 
Haploid Embryonic Stem Cells and the Dominance of Recessive Traits
Mesp1 at the Heart of Mesoderm Lineage Specification
Rejuvenation: It’s in Our Blood
Cellular Alchemy and the Golden Age of Reprogramming
Modeling Brain Disease in a Dish: Really?
Young at Heart Cell Volume 153, Issue 4, Pages (May 2013)
Stems Cells and the Pathways to Aging and Cancer
Presentation transcript:

Programming and Reprogramming Cellular Age in the Era of Induced Pluripotency  Lorenz Studer, Elsa Vera, Daniela Cornacchia  Cell Stem Cell  Volume 16, Issue 6, Pages 591-600 (June 2015) DOI: 10.1016/j.stem.2015.05.004 Copyright © 2015 Elsevier Inc. Terms and Conditions

Figure 1 Traditional Approaches and Cellular Hallmarks of Aging (A) Screens in model organisms were crucial for the identification of signaling pathways that increase organismal lifespan. Such pathways include insulin/IGF1 signaling (IIS) and nutrient-sensing pathways among others. An alternative strategy is focused on the identification of genomic determinants of longevity through the study of exceptionally long-lived or short-lived animals. Organisms with unusually long lifespans include the naked mole rat, the bowhead whale, or the Brandt’s bat. Examples of short-lived animals of interest in aging research include the killifish and hydra. (B) Heterochronic parabiosis has become an important tool for the discovery of systemic factors capable of restoring a youthful regenerative capacity in aged tissues through shared blood circulation with a young animal. Blood-borne factors capable of mediating the effects of parabiosis have been identified including factors promoting skeletal muscle, blood vessel, brain, and heart regeneration. (C) An additional approach to study aging represents the investigation of cellular alterations that accompany organismal aging. Cellular hallmarks of age that can be readily measured can affect virtually any compartment of the cell. Additional hallmarks such as altered intracellular communication and defects in nutrient sensing are not shown here because they are difficult to quantify in vitro. Cell Stem Cell 2015 16, 591-600DOI: (10.1016/j.stem.2015.05.004) Copyright © 2015 Elsevier Inc. Terms and Conditions

Figure 2 Phenotypic Rejuvenation during iPSC Induction (A) Fibroblasts from an aged donor can be reprogrammed back to pluripotency and further differentiated into iPSC-derived fibroblasts. During this process, many cellular aging hallmarks are reset to a young-like state ranging from cellular senescence to nuclear morphology. (B) Legend for each of the age-related hallmarks reset during reprogramming. Cell Stem Cell 2015 16, 591-600DOI: (10.1016/j.stem.2015.05.004) Copyright © 2015 Elsevier Inc. Terms and Conditions

Figure 3 Manipulating Cellular Age in Pluripotent-Derived Cells (A) The three stages of developmental timing: cell-fate specification, maturation, and aging. In vitro differentiation of hPSC to neuronal lineages follows a temporal program reminiscent of brain development in vivo. PSC-derived neurons commonly represent immature fetal-stage cells, and novel tools are under development to improve functional maturation and trigger expression of age-related markers in pluripotent-derived cells. (B) Modeling late-onset disorders in patient-specific cells may require the induction of stress or age-related markers to uncover disease related phenotypes. Most studies so far focused on challenging cells with toxic stressors. We postulate that such paradigms are suitable to reveal disease-related susceptibility phenotypes that reflect early biochemical changes in the disease process “early biochemical.” We propose “inducing aging” as an alternative strategy to “inducing stress” that may reveal additional disease phenotypes. As opposed to inducing stress, strategies aimed at inducing age are thought to trigger expression of the various molecular hallmarks of age. Induced aging strategies draw inspiration from naturally occurring mutations known to cause premature aging, such as the recently reported method based on the expression of progerin. An alternative strategy that has not yet been implemented for human disease modeling could involve the direct manipulation of one or a subset of molecular hallmarks of aging with the goal of eliciting cellular age in a more physiological manner. Cell Stem Cell 2015 16, 591-600DOI: (10.1016/j.stem.2015.05.004) Copyright © 2015 Elsevier Inc. Terms and Conditions