Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where F. Brad Johnson is active.

Publication


Featured researches published by F. Brad Johnson.


Nature | 2009

Histone H4 lysine 16 acetylation regulates cellular lifespan.

Weiwei Dang; Kristan K. Steffen; Rocco Perry; Jean Dorsey; F. Brad Johnson; Ali Shilatifard; Matt Kaeberlein; Brian K. Kennedy; Shelley L. Berger

Cells undergoing developmental processes are characterized by persistent non-genetic alterations in chromatin, termed epigenetic changes, represented by distinct patterns of DNA methylation and histone post-translational modifications. Sirtuins, a group of conserved NAD+-dependent deacetylases or ADP-ribosyltransferases, promote longevity in diverse organisms; however, their molecular mechanisms in ageing regulation remain poorly understood. Yeast Sir2, the first member of the family to be found, establishes and maintains chromatin silencing by removing histone H4 lysine 16 acetylation and bringing in other silencing proteins. Here we report an age-associated decrease in Sir2 protein abundance accompanied by an increase in H4 lysine 16 acetylation and loss of histones at specific subtelomeric regions in replicatively old yeast cells, which results in compromised transcriptional silencing at these loci. Antagonizing activities of Sir2 and Sas2, a histone acetyltransferase, regulate the replicative lifespan through histone H4 lysine 16 at subtelomeric regions. This pathway, distinct from existing ageing models for yeast, may represent an evolutionarily conserved function of sirtuins in regulation of replicative ageing by maintenance of intact telomeric chromatin.


Molecular and Cellular Biology | 2004

Telomere Shortening Exposes Functions for the Mouse Werner and Bloom Syndrome Genes

Xiaobing Du; Johnny Shen; Nishan Kugan; Emma E. Furth; David B. Lombard; Catherine Cheung; Sally Pak; Guangbin Luo; Robert J. Pignolo; Ronald A. DePinho; Leonard Guarente; F. Brad Johnson

ABSTRACT The Werner and Bloom syndromes are caused by loss-of-function mutations in WRN and BLM, respectively, which encode the RecQ family DNA helicases WRN and BLM, respectively. Persons with Werner syndrome displays premature aging of the skin, vasculature, reproductive system, and bone, and those with Bloom syndrome display more limited features of aging, including premature menopause; both syndromes involve genome instability and increased cancer. The proteins participate in recombinational repair of stalled replication forks or DNA breaks, but the precise functions of the proteins that prevent rapid aging are unknown. Accumulating evidence points to telomeres as targets of WRN and BLM, but the importance in vivo of the proteins in telomere biology has not been tested. We show that Wrn and Blm mutations each accentuate pathology in later-generation mice lacking the telomerase RNA template Terc, including acceleration of phenotypes characteristic of latest-generation Terc mutants. Furthermore, pathology not observed in Terc mutants but similar to that observed in Werner syndrome and Bloom syndrome, such as bone loss, was observed. The pathology was accompanied by enhanced telomere dysfunction, including end-to-end chromosome fusions and greater loss of telomere repeat DNA compared with Terc mutants. These findings indicate that telomere dysfunction may contribute to the pathogenesis of Werner syndrome and Bloom syndrome.


Nucleic Acids Research | 2008

Genomic distribution and functional analyses of potential G-quadruplex-forming sequences in Saccharomyces cerevisiae.

Steve G. Hershman; Qijun Chen; Julia Y. Lee; Marina L. Kozak; Peng Yue; Li-San Wang; F. Brad Johnson

Although well studied in vitro, the in vivo functions of G-quadruplexes (G4-DNA and G4-RNA) are only beginning to be defined. Recent studies have demonstrated enrichment for sequences with intramolecular G-quadruplex forming potential (QFP) in transcriptional promoters of humans, chickens and bacteria. Here we survey the yeast genome for QFP sequences and similarly find strong enrichment for these sequences in upstream promoter regions, as well as weaker but significant enrichment in open reading frames (ORFs). Further, four findings are consistent with roles for QFP sequences in transcriptional regulation. First, QFP is correlated with upstream promoter regions with low histone occupancy. Second, treatment of cells with N-methyl mesoporphyrin IX (NMM), which binds G-quadruplexes selectively in vitro, causes significant upregulation of loci with QFP-possessing promoters or ORFs. NMM also causes downregulation of loci connected with the function of the ribosomal DNA (rDNA), which itself has high QFP. Third, ORFs with QFP are selectively downregulated in sgs1 mutants that lack the G4-DNA-unwinding helicase Sgs1p. Fourth, a screen for yeast mutants that enhance or suppress growth inhibition by NMM revealed enrichment for chromatin and transcriptional regulators, as well as telomere maintenance factors. These findings raise the possibility that QFP sequences form bona fide G-quadruplexes in vivo and thus regulate transcription.


Nucleic Acids Research | 2010

Altered gene expression in the Werner and Bloom syndromes is associated with sequences having G-quadruplex forming potential

Jay E. Johnson; Kajia Cao; Paul Ryvkin; Li-San Wang; F. Brad Johnson

The human Werner and Bloom syndromes (WS and BS) are caused by deficiencies in the WRN and BLM RecQ helicases, respectively. WRN, BLM and their Saccharomyces cerevisiae homologue Sgs1, are particularly active in vitro in unwinding G-quadruplex DNA (G4-DNA), a family of non-canonical nucleic acid structures formed by certain G-rich sequences. Recently, mRNA levels from loci containing potential G-quadruplex-forming sequences (PQS) were found to be preferentially altered in sgs1Δ mutants, suggesting that G4-DNA targeting by Sgs1 directly affects gene expression. Here, we extend these findings to human cells. Using microarrays to measure mRNAs obtained from human fibroblasts deficient for various RecQ family helicases, we observe significant associations between loci that are upregulated in WS or BS cells and loci that have PQS. No such PQS associations were observed for control expression datasets, however. Furthermore, upregulated genes in WS and BS showed no or dramatically reduced associations with sequences similar to PQS but that have considerably reduced potential to form intramolecular G4-DNA. These findings indicate that, like Sgs1, WRN and BLM can regulate transcription globally by targeting G4-DNA.


Aging Cell | 2008

Defects in telomere maintenance molecules impair osteoblast differentiation and promote osteoporosis.

Robert J. Pignolo; Robin K. Suda; Emily A. McMillan; Johnny Shen; Seoung-Hoon Lee; Yongwon Choi; Alexander C. Wright; F. Brad Johnson

Osteoporosis and the associated risk of fracture are major clinical challenges in the elderly. Telomeres shorten with age in most human tissues, including bone, and because telomere shortening is a cause of cellular replicative senescence or apoptosis in cultured cells, including mesenchymal stem cells (MSCs) and osteoblasts, it is hypothesized that telomere shortening contributes to the aging of bone. Osteoporosis is common in the Werner (Wrn) and dyskeratosis congenita premature aging syndromes, which are characterized by telomere dysfunction. One of the targets of the Wrn helicase is telomeric DNA, but the long telomeres and abundant telomerase in mice minimize the need for Wrn at telomeres, and thus Wrn knockout mice are relatively healthy. In a model of accelerated aging that combines the Wrn mutation with the shortened telomeres of telomerase (Terc) knockout mice, synthetic defects in proliferative tissues result. Here, we demonstrate that deficiencies in Wrn−/– Terc−/– mutant mice cause a low bone mass phenotype, and that age‐related osteoporosis is the result of impaired osteoblast differentiation in the context of intact osteoclast differentiation. Further, MSCs from single and Wrn−/– Terc−/– double mutant mice have a reduced in vitro lifespan and display impaired osteogenic potential concomitant with characteristics of premature senescence. These data provide evidence that replicative aging of osteoblast precursors is an important mechanism of senile osteoporosis.


Cancer immunology research | 2015

Identification of chimeric antigen receptors that mediate constitutive or inducible proliferation of T cells.

Matthew J. Frigault; Jihyun Lee; Maria Ciocca Basil; Carmine Carpenito; Shinichiro Motohashi; John Scholler; Omkar U. Kawalekar; Sonia Guedan; Shannon E. McGettigan; Avery D. Posey; Sonny Ang; Laurence J.N. Cooper; Jesse Platt; F. Brad Johnson; Chrystal M. Paulos; Yangbing Zhao; Michael Kalos; Michael C. Milone; Carl H. June

Frigault, Lee, and colleagues compared chimeric antigen receptors (CAR) encoding signaling domains comprising CD28, ICOS, and 4-1BB and found that some CD28 CAR-T cells have antigen-independent constitutive proliferation and cytokine secretion when highly expressed, leading to inferior antitumor effects. This study compared second-generation chimeric antigen receptors (CAR) encoding signaling domains composed of CD28, ICOS, and 4-1BB (TNFRSF9). Here, we report that certain CARs endow T cells with the ability to undergo long-term autonomous proliferation. Transduction of primary human T cells with lentiviral vectors encoding some of the CARs resulted in sustained proliferation for up to 3 months following a single stimulation through the T-cell receptor (TCR). Sustained numeric expansion was independent of cognate antigen and did not require the addition of exogenous cytokines or feeder cells after a single stimulation of the TCR and CD28. Results from gene array and functional assays linked sustained cytokine secretion and expression of T-bet (TBX21), EOMES, and GATA-3 to the effect. Sustained expression of the endogenous IL2 locus has not been reported in primary T cells. Sustained proliferation was dependent on CAR structure and high expression, the latter of which was necessary but not sufficient. The mechanism involves constitutive signaling through NF-κB, AKT, ERK, and NFAT. The propagated CAR T cells retained a diverse TCR repertoire, and cellular transformation was not observed. The CARs with a constitutive growth phenotype displayed inferior antitumor effects and engraftment in vivo. Therefore, the design of CARs that have a nonconstitutive growth phenotype may be a strategy to improve efficacy and engraftment of CAR T cells. The identification of CARs that confer constitutive or nonconstitutive growth patterns may explain observations that CAR T cells have differential survival patterns in clinical trials. Cancer Immunol Res; 3(4); 356–67. ©2015 AACR.


Alzheimers & Dementia | 2009

Comparisons of telomere lengths in peripheral blood and cerebellum in Alzheimer's disease

J. Nicholas Lukens; Vivianna M. Van Deerlin; Christopher M. Clark; Sharon X. Xie; F. Brad Johnson

Alzheimers disease (AD) patients have been reported to have shorter telomeres in peripheral blood leukocytes (PBLs) than age‐matched control subjects. However, it is unclear if PBL telomere length reflects brain telomere length, which might play a more direct role in AD pathogenesis. We examined the correlation between PBL and cerebellum telomere length in AD patients, and compared telomere lengths in cerebella from individuals with AD versus age‐matched control subjects.


Cell Reports | 2015

DNA-Damage-Induced Type I Interferon Promotes Senescence and Inhibits Stem Cell Function

Qiujing Yu; Yuliya V. Katlinskaya; Christopher J. Carbone; Bin Zhao; Kanstantsin V. Katlinski; Hui Zheng; Manti Guha; Ning Li; Qijun Chen; Ting Yang; Christopher J. Lengner; Roger A. Greenberg; F. Brad Johnson; Serge Y. Fuchs

Expression of type I interferons (IFNs) can be induced by DNA-damaging agents, but the mechanisms and significance of this regulation are not completely understood. We found that the transcription factor IRF3, activated in an ATM-IKKα/β-dependent manner, stimulates cell-autonomous IFN-β expression in response to double-stranded DNA breaks. Cells and tissues with accumulating DNA damage produce endogenous IFN-β and stimulate IFN signaling in vitro and in vivo. In turn, IFN acts to amplify DNA-damage responses, activate the p53 pathway, promote senescence, and inhibit stem cell function in response to telomere shortening. Inactivation of the IFN pathway abrogates the development of diverse progeric phenotypes and extends the lifespan of Terc knockout mice. These data identify DNA-damage-response-induced IFN signaling as a critical mechanism that links accumulating DNA damage with senescence and premature aging.


Nature Structural & Molecular Biology | 2011

Rudimentary G-quadruplex–based telomere capping in Saccharomyces cerevisiae

Jasmine S. Smith; Qijun Chen; Liliya A. Yatsunyk; John M. Nicoludis; Mark S Garcia; Ramon Kranaster; Shankar Balasubramanian; David Monchaud; Marie-Paule Teulade-Fichou; Lara K. Abramowitz; David C. Schultz; F. Brad Johnson

Telomere capping conceals chromosome ends from exonucleases and checkpoints, but the full range of capping mechanisms is not well defined. Telomeres have the potential to form G-quadruplex (G4) DNA, although evidence for telomere G4 DNA function in vivo is limited. In budding yeast, capping requires the Cdc13 protein and is lost at nonpermissive temperatures in cdc13-1 mutants. Here, we use several independent G4 DNA–stabilizing treatments to suppress cdc13-1 capping defects. These include overexpression of three different G4 DNA binding proteins, loss of the G4 DNA unwinding helicase Sgs1, or treatment with small molecule G4 DNA ligands. In vitro, we show that protein-bound G4 DNA at a 3′ overhang inhibits 5′→3′ resection of a paired strand by exonuclease I. These findings demonstrate that, at least in the absence of full natural capping, G4 DNA can play a positive role at telomeres in vivo.


Biochimie | 2008

In vivo veritas: using yeast to probe the biological functions of G-quadruplexes

Jay E. Johnson; Jasmine S. Smith; Marina L. Kozak; F. Brad Johnson

Certain guanine-rich sequences are capable of forming higher order structures known as G-quadruplexes. Moreover, particular genomic regions in a number of highly divergent organisms are enriched for such sequences, raising the possibility that G-quadruplexes form in vivo and affect cellular processes. While G-quadruplexes have been rigorously studied in vitro, whether these structures actually form in vivo and what their roles might be in the context of the cell have remained largely unanswered questions. Recent studies suggest that G-quadruplexes participate in the regulation of such varied processes as telomere maintenance, transcriptional regulation and ribosome biogenesis. Here we review studies aimed at elucidating the in vivo functions of quadruplex structures, with a particular focus on findings in yeast. In addition, we discuss the utility of yeast model systems in the study of the cellular roles of G-quadruplexes.

Collaboration


Dive into the F. Brad Johnson's collaboration.

Top Co-Authors

Avatar

Qijun Chen

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Shelley L. Berger

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Li-San Wang

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Jasmine S. Smith

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Brian D. Gregory

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kajia Cao

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Leonard Guarente

Massachusetts Institute of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge