Network


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

Hotspot


Dive into the research topics where R. Keaney Rathbun is active.

Publication


Featured researches published by R. Keaney Rathbun.


Molecular and Cellular Biology | 2000

The Fanconi anemia protein FANCC binds to and facilitates the activation of STAT1 by gamma interferon and hematopoietic growth factors.

Qishen Pang; Sara R. Fagerlie; Tracy A. Christianson; Winifred Keeble; Greg Faulkner; Jane Diaz; R. Keaney Rathbun; Grover C. Bagby

ABSTRACT Hematopoietic progenitor cells from Fanconi anemia (FA) group C (FA-C) patients display hypersensitivity to the apoptotic effects of gamma interferon (IFN-γ) and constitutively express a variety of IFN-dependent genes. Paradoxically, however, STAT1 activation is suppressed in IFN-stimulated FA cells, an abnormality corrected by transduction of normal FANCC cDNA. We therefore sought to define the specific role of FANCC protein in signal transduction through receptors that activate STAT1. Expression and phosphorylation of IFN-γ receptor α chain (IFN-γRα) and JAK1 and JAK2 tyrosine kinases were equivalent in both normal and FA-C cells. However, in coimmunoprecipitation experiments STAT1 did not dock at the IFN-γR of FA-C cells, an abnormality corrected by transduction of the FANCC gene. In addition, glutathione S-transferase fusion genes encoding normal FANCC but not a mutant FANCC bearing an inactivating point mutation (L554P) bound to STAT1 in lysates of IFN-γ-stimulated B cells and IFN-, granulocyte-macrophage colony-stimulating factor- and stem cell factor-stimulated MO7e cells. Kinetic studies revealed that the initial binding of FANCC was to nonphosphorylated STAT1 but that subsequently the complex moved to the receptor docking site, at which point STAT1 became phosphorylated. The STAT1 phosphorylation defect in FA-C cells was functionally significant in that IFN induction of IFN response factor 1 was suppressed and STAT1-DNA complexes were not detected in nuclear extracts of FA-C cells. We also determined that the IFN-γ hypersensitivity of FA-C hematopoietic progenitor cells does not derive from STAT1 activation defects because granulocyte-macrophage CFU and erythroid burst-forming units from STAT1−/− mice were resistant to IFN-γ. However, BFU-E responses to SCF and erythropoietin were suppressed in STAT−/− mice. Consequently, because the FANCC protein is involved in the activation of STAT1 through receptors for at least three hematopoietic growth and survival factor molecules, we reason that FA-C hematopoietic cells are excessively apoptotic because of an imbalance between survival cues (owing to a failure of STAT1 activation in FA-C cells) and apoptotic and mitogenic inhibitory cues (constitutively activated in FA-C cells in a STAT1-independent fashion).


Blood | 2010

Fancd2−/− mice have hematopoietic defects that can be partially corrected by resveratrol

Qing Shuo Zhang; Laura Marquez-Loza; Laura Eaton; Andrew W. Duncan; Devorah C. Goldman; Praveen Anur; Kevin Watanabe-Smith; R. Keaney Rathbun; William H. Fleming; Grover C. Bagby; Markus Grompe

Progressive bone marrow failure is a major cause of morbidity and mortality in human Fanconi Anemia patients. In an effort to develop a Fanconi Anemia murine model to study bone marrow failure, we found that Fancd2(-/-) mice have readily measurable hematopoietic defects. Fancd2 deficiency was associated with a significant decline in the size of the c-Kit(+)Sca-1(+)Lineage(-) (KSL) pool and reduced stem cell repopulation and spleen colony-forming capacity. Fancd2(-/-) KSL cells showed an abnormal cell cycle status and loss of quiescence. In addition, the supportive function of the marrow microenvironment was compromised in Fancd2(-/-) mice. Treatment with Sirt1-mimetic and the antioxidant drug, resveratrol, maintained Fancd2(-/-) KSL cells in quiescence, improved the marrow microenvironment, partially corrected the abnormal cell cycle status, and significantly improved the spleen colony-forming capacity of Fancd2(-/-) bone marrow cells. We conclude that Fancd2(-/-) mice have readily quantifiable hematopoietic defects, and that this model is well suited for pharmacologic screening studies.


Blood | 2009

TLR8-dependent TNF-α overexpression in Fanconi anemia group C cells

Scott M. Vanderwerf; Johanna Svahn; Susan B. Olson; R. Keaney Rathbun; Christina A. Harrington; Jane Yates; Winifred Keeble; David C. Anderson; Praveen Anur; Noemi F. Pereira; Daniela Pilonetto; Ricardo Pasquini; Grover C. Bagby

Tumor necrosis factor alpha (TNF-alpha) production is abnormally high in Fanconi anemia (FA) cells and contributes to the hematopoietic defects seen in FA complementation group C-deficient (Fancc(-/-)) mice. Applying gene expression microarray and proteomic methods to studies on FANCC-deficient cells we found that genes encoding proteins directly involved in ubiquitinylation are overrepresented in the signature of FA bone marrow cells and that ubiquitinylation profiles of FA-C and complemented cells were substantially different. Finding that Toll-like receptor 8 (TLR8) was one of the proteins ubiquitinylated only in mutant cells, we confirmed that TLR8 (or a TLR8-associated protein) is ubiquitinylated in mutant FA-C cells and that TNF-alpha production in mutant cells depended upon TLR8 and the canonical downstream signaling intermediates interleukin 1 receptor-associated kinase (IRAK) and IkappaB kinase-alpha/beta. FANCC-deficient THP-1 cells and macrophages from Fancc(-/-) mice overexpressed TNF-alpha in response to TLR8 agonists but not other TLR agonists. Ectopically expressed FANCC point mutants were capable of fully complementing the mitomycin-C hypersensitivity phenotype of FA-C cells but did not suppress TNF-alpha overproduction. In conclusion, FANCC suppresses TNF-alpha production in mononuclear phagocytes by suppressing TLR8 activity and this particular function of FANCC is independent of its function in protecting the genome from cross-linking agents.


Cancer Research | 2006

Cytogenetic Instability in Ovarian Epithelial Cells from Women at Risk of Ovarian Cancer

Tanja Pejovic; Jane Yates; Hong Y. Liu; Laura E. Hays; Yassmine Akkari; Yumi Torimaru; Winifred Keeble; R. Keaney Rathbun; William H. Rodgers; Allen E. Bale; Najim Ameziane; C. Michael Zwaan; Abdellatif Errami; Philippe Thuillier; Fabio Cappuccini; Susan B. Olson; Joanna M. Cain; Grover C. Bagby

Fanconi anemia is an inherited cancer predisposition disease characterized by cytogenetic and cellular hypersensitivity to cross-linking agents. Seeking evidence of Fanconi anemia protein dysfunction in women at risk of ovarian cancer, we screened ovarian surface epithelial cells from 25 primary cultures established from 22 patients using cross-linker hypersensitivity assays. Samples were obtained from (a) women at high risk for ovarian cancer with histologically normal ovaries, (b) ovarian cancer patients, and (c) a control group with no family history of breast or ovarian cancer. In chromosomal breakage assays, all control cells were mitomycin C (MMC) resistant, but eight samples (five of the six high-risk and three of the eight ovarian cancer) were hypersensitive. Lymphocytes from all eight patients were MMC resistant. Only one of the eight patients had a BRCA1 germ-line mutation and none had BRCA2 mutations, but FANCD2 was reduced in five of the eight. Ectopic expression of normal FANCD2 cDNA increased FANCD2 protein and induced MMC resistance in both hypersensitive lines tested. No FANCD2 coding region or promoter mutations were found, and there was no genomic loss or promoter methylation in any Fanconi anemia genes. Therefore, in high-risk women with no BRCA1 or BRCA2 mutations, tissue-restricted hypersensitivity to cross-linking agents is a frequent finding, and chromosomal breakage responses to MMC may be a sensitive screening strategy because cytogenetic instability identified in this way antedates the onset of carcinoma. Inherited mutations that result in tissue-specific FANCD2 gene suppression may represent a cause of familial ovarian cancer.


Stem cell reports | 2015

Oxymetholone Therapy of Fanconi Anemia Suppresses Osteopontin Transcription and Induces Hematopoietic Stem Cell Cycling

Qing Shuo Zhang; Eric Benedetti; Matthew Deater; Kathryn Schubert; Angela Major; Carl Pelz; Soren Impey; Laura Marquez-Loza; R. Keaney Rathbun; Shigeaki Kato; Grover C. Bagby; Markus Grompe

Summary Androgens are widely used for treating Fanconi anemia (FA) and other human bone marrow failure syndromes, but their mode of action remains incompletely understood. Aged Fancd2−/− mice were used to assess the therapeutic efficacy of oxymetholone (OXM) and its mechanism of action. Eighteen-month-old Fancd2−/− mice recapitulated key human FA phenotypes, including reduced bone marrow cellularity, red cell macrocytosis, and peripheral pancytopenia. As in humans, chronic OXM treatment significantly improved these hematological parameters and stimulated the proliferation of hematopoietic stem and progenitor cells. RNA-Seq analysis implicated downregulation of osteopontin as an important potential mechanism for the drug’s action. Consistent with the increased stem cell proliferation, competitive repopulation assays demonstrated that chronic OXM therapy eventually resulted in stem cell exhaustion. These results expand our knowledge of the regulation of hematopoietic stem cell proliferation and have direct clinical implications for the treatment of bone marrow failure.


Journal of Leukocyte Biology | 2016

Cytokine overproduction and crosslinker hypersensitivity are unlinked in Fanconi anemia macrophages

Michael Garbati; Laura E. Hays; R. Keaney Rathbun; Nathaniel Jillette; Kathy Chin; Muhsen Al-Dhalimy; Anupriya Agarwal; Amy Hanlon Newell; Susan B. Olson; Grover C. Bagby

The Fanconi anemia proteins participate in a canonical pathway that repairs cross‐linking agent‐induced DNA damage. Cells with inactivated Fanconi anemia genes are universally hypersensitive to such agents. Fanconi anemia‐deficient hematopoietic stem cells are also hypersensitive to inflammatory cytokines, and, as importantly, Fanconi anemia macrophages overproduce such cytokines in response to TLR4 and TLR7/8 agonists. We questioned whether TLR‐induced DNA damage is the primary cause of aberrantly regulated cytokine production in Fanconi anemia macrophages by quantifying TLR agonist‐induced TNF‐α production, DNA strand breaks, crosslinker‐induced chromosomal breakage, and Fanconi anemia core complex function in Fanconi anemia complementation group C‐deficient human and murine macrophages. Although both M1 and M2 polarized Fanconi anemia cells were predictably hypersensitive to mitomycin C, only M1 macrophages overproduced TNF‐α in response to TLR‐activating signals. DNA damaging agents alone did not induce TNF‐α production in the absence of TLR agonists in wild‐type or Fanconi anemia macrophages, and mitomycin C did not enhance TLR responses in either normal or Fanconi anemia cells. TLR4 and TLR7/8 activation induced cytokine overproduction in Fanconi anemia macrophages. Also, although TLR4 activation was associated with induced double strand breaks, TLR7/8 activation was not. That DNA strand breaks and chromosome breaks are neither necessary nor sufficient to account for the overproduction of inflammatory cytokines by Fanconi anemia cells suggests that noncanonical anti‐inflammatory functions of Fanconi anemia complementation group C contribute to the aberrant macrophage phenotype and suggests that suppression of macrophage/TLR hyperreactivity might prevent cytokine‐induced stem cell attrition in Fanconi anemia.


Experimental Hematology | 2000

Fanconi anemia and apoptosis: The fancc gene product functions downstream of caspase 3

R. Keaney Rathbun; Tracy A. Christianson; Paul S. Koh; Gary Jones; Winifred Keeble; Grover C. Bagby

Abstract Hematopoietic progenitor cells (HPC) from mice and children with type C Fanconi anemia (FA-C) are hypersensitive to mitotic inhibitory factors, including interferon-γ (IFN-γ) and tumor necrosis factor-α (TNFα)(Blood, 90:974,1997). We tested the hypothesis that IFN/fasL induced programmed cell death in FA-C cells involves the ordered activation of specific caspase molecules. In immunoblot experiments, the FA-C lymphoblastoid cell line HSC536N treated with an agonistic fas antibody (100 ng/ml) and IFN-γ(1 ng/ml) contained activated caspase 3, 6, and 7 (but not 1, 2, 4, or 10) by 60 minutes and PARP cleavage products by 180 minutes. Caspase 3 activation by immunoblot and flow cytometric analysis was identical in FA-C cells and FA cells complemented by FANCC cDNA transfer even though the latter cells were less apoptotic. The apoptotic effects of fas agonists in IFN-γ-treated human FA-C CD34 + cells, FA-C murine progenitor cells, and EBV-transformed lymphoblasts from a child with FA of the C type, were blocked when the cells were pretreated any one of three inhibitors of caspase 3 protease. Caspase 1 inhibitors did not block the effect of IFN and fas ligand and caspase 1 inhibition did not prevent caspase 3 cleavage but caspase 8 inhibition did block caspase 3 activation. We conclude that in IFN-primed FA-C cells, fas -induced apoptosis involves the activation of caspase 8, which controls activation of caspase 3 family members but that the function of FANCC in suppressing normal apoptotic responses is downstream of caspase 3.


Blood | 1997

Inactivation of the Fanconi Anemia Group C Gene Augments Interferon-γ–Induced Apoptotic Responses in Hematopoietic Cells

R. Keaney Rathbun; Gregory R. Faulkner; Marika H. Ostroski; Tracy A. Christianson; Grant Hughes; Gary Jones; Robert W. Cahn; Richard T. Maziarz; Gordon Royle; Winifred Keeble; Michael C. Heinrich; Markus Grompe; Paula A. Tower; Grover C. Bagby


Blood | 2000

Interferon-γ-induced apoptotic responses of Fanconi anemia group C hematopoietic progenitor cells involve caspase 8-dependent activation of caspase 3 family members

R. Keaney Rathbun; Tracy A. Christianson; Gregory R. Faulkner; Gary Jones; Winifred Keeble; Michael O'Dwyer; Grover C. Bagby


Experimental Parasitology | 2006

Evaluation and lead optimization of anti-malarial acridones

Rolf W. Winter; Jane Xu Kelly; Martin J. Smilkstein; Rozalia A. Dodean; Grover C. Bagby; R. Keaney Rathbun; David J. Hinrichs; Michael K. Riscoe

Collaboration


Dive into the R. Keaney Rathbun's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge