Network


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

Hotspot


Dive into the research topics where Katherine J. Ladner is active.

Publication


Featured researches published by Katherine J. Ladner.


Cancer Cell | 2008

NF-κB–YY1–miR-29 Regulatory Circuitry in Skeletal Myogenesis and Rhabdomyosarcoma

Huating Wang; Ramiro Garzon; Hao Sun; Katherine J. Ladner; Ravi K. Singh; Jason M. Dahlman; Alfred S.L. Cheng; Brett M. Hall; Stephen J. Qualman; Dawn S. Chandler; Carlo M. Croce; Denis C. Guttridge

Studies support the importance of microRNAs in physiological and pathological processes. Here we describe the regulation and function of miR-29 in myogenesis and rhabdomyosarcoma (RMS). Results demonstrate that in myoblasts, miR-29 is repressed by NF-kappaB acting through YY1 and the Polycomb group. During myogenesis, NF-kappaB and YY1 downregulation causes derepression of miR-29, which in turn accelerates differentiation by targeting its repressor YY1. However, in RMS cells and primary tumors that possess impaired differentiation, miR-29 is epigenetically silenced by an activated NF-kappaB-YY1 pathway. Reconstitution of miR-29 in RMS in mice inhibits tumor growth and stimulates differentiation, suggesting that miR-29 acts as a tumor suppressor through its promyogenic function. Together, these results identify a NF-kappaB-YY1-miR-29 regulatory circuit whose disruption may contribute to RMS.


Journal of Clinical Investigation | 2004

Cancer cachexia is regulated by selective targeting of skeletal muscle gene products

Swarnali Acharyya; Katherine J. Ladner; Lori L. Nelsen; Jeffrey S. Damrauer; Peter J. Reiser; Steven Swoap; Denis C. Guttridge

Cachexia is a syndrome characterized by wasting of skeletal muscle and contributes to nearly one-third of all cancer deaths. Cytokines and tumor factors mediate wasting by suppressing muscle gene products, but exactly which products are targeted by these cachectic factors is not well understood. Because of their functional relevance to muscle architecture, such targets are presumed to represent myofibrillar proteins, but whether these proteins are regulated in a general or a selective manner is also unclear. Here we demonstrate, using in vitro and in vivo models of muscle wasting, that cachectic factors are remarkably selective in targeting myosin heavy chain. In myotubes and mouse muscles, TNF-alpha plus IFN-gamma strongly reduced myosin expression through an RNA-dependent mechanism. Likewise, colon-26 tumors in mice caused the selective reduction of this myofibrillar protein, and this reduction correlated with wasting. Under these conditions, however, loss of myosin was associated with the ubiquitin-dependent proteasome pathway, which suggests that mechanisms used to regulate the expression of muscle proteins may be cachectic factor specific. These results shed new light on cancer cachexia by revealing that wasting does not result from a general downregulation of muscle proteins but rather is highly selective as to which proteins are targeted during the wasting state.


Annals of Neurology | 2011

DUX4, a candidate gene for facioscapulohumeral muscular dystrophy, causes p53-dependent myopathy in vivo

Lindsay M. Wallace; Sara E. Garwick; Wenyan Mei; Alexandra Belayew; Frédérique Coppée; Katherine J. Ladner; Denis C. Guttridge; Jing Yang; Scott Q. Harper

Facioscapulohumeral muscular dystrophy (FSHD) is associated with D4Z4 repeat contraction on human chromosome 4q35. This genetic lesion does not result in complete loss or mutation of any gene. Consequently, the pathogenic mechanisms underlying FSHD have been difficult to discern. In leading FSHD pathogenesis models, D4Z4 contractions are proposed to cause epigenetic changes, which ultimately increase expression of genes with myopathic potential. Although no gene has been conclusively linked to FSHD development, recent evidence supports a role for the D4Z4‐encoded DUX4 gene in FSHD. In this study, our objective was to test the in vivo myopathic potential of DUX4.


Journal of Clinical Investigation | 2013

NF-κB-mediated Pax7 dysregulation in the muscle microenvironment promotes cancer cachexia

Wei A. He; Emanuele Berardi; Veronica Cardillo; Swarnali Acharyya; Paola Aulino; Jennifer Thomas-Ahner; Jingxin Wang; Mark Bloomston; Peter Muscarella; Peter Nau; Nilay Shah; Matthew E.R. Butchbach; Katherine J. Ladner; Sergio Adamo; Michael A. Rudnicki; Charles Keller; Dario Coletti; Federica Montanaro; Denis C. Guttridge

Cachexia is a debilitating condition characterized by extreme skeletal muscle wasting that contributes significantly to morbidity and mortality. Efforts to elucidate the underlying mechanisms of muscle loss have predominantly focused on events intrinsic to the myofiber. In contrast, less regard has been given to potential contributory factors outside the fiber within the muscle microenvironment. In tumor-bearing mice and patients with pancreatic cancer, we found that cachexia was associated with a type of muscle damage resulting in activation of both satellite and nonsatellite muscle progenitor cells. These muscle progenitors committed to a myogenic program, but were inhibited from completing differentiation by an event linked with persistent expression of the self-renewing factor Pax7. Overexpression of Pax7 was sufficient to induce atrophy in normal muscle, while under tumor conditions, the reduction of Pax7 or exogenous addition of its downstream target, MyoD, reversed wasting by restoring cell differentiation and fusion with injured fibers. Furthermore, Pax7 was induced by serum factors from cachectic mice and patients, in an NF-κB-dependent manner, both in vitro and in vivo. Together, these results suggest that Pax7 responds to NF-κB by impairing the regenerative capacity of myogenic cells in the muscle microenvironment to drive muscle wasting in cancer.


PLOS ONE | 2010

TNF Inhibits Notch-1 in Skeletal Muscle Cells by Ezh2 and DNA Methylation Mediated Repression: Implications in Duchenne Muscular Dystrophy

Swarnali Acharyya; Sudarshana M. Sharma; Alfred S.L. Cheng; Katherine J. Ladner; Wei He; William Kline; Huating Wang; Michael C. Ostrowski; Tim H M Huang; Denis C. Guttridge

Background Classical NF-κB signaling functions as a negative regulator of skeletal myogenesis through potentially multiple mechanisms. The inhibitory actions of TNFα on skeletal muscle differentiation are mediated in part through sustained NF-κB activity. In dystrophic muscles, NF-κB activity is compartmentalized to myofibers to inhibit regeneration by limiting the number of myogenic progenitor cells. This regulation coincides with elevated levels of muscle derived TNFα that is also under IKKβ and NF-κB control. Methodology/Principal Findings Based on these findings we speculated that in DMD, TNFα secreted from myotubes inhibits regeneration by directly acting on satellite cells. Analysis of several satellite cell regulators revealed that TNFα is capable of inhibiting Notch-1 in satellite cells and C2C12 myoblasts, which was also found to be dependent on NF-κB. Notch-1 inhibition occurred at the mRNA level suggesting a transcriptional repression mechanism. Unlike its classical mode of action, TNFα stimulated the recruitment of Ezh2 and Dnmt-3b to coordinate histone and DNA methylation, respectively. Dnmt-3b recruitment was dependent on Ezh2. Conclusions/Significance We propose that in dystrophic muscles, elevated levels of TNFα and NF-κB inhibit the regenerative potential of satellite cells via epigenetic silencing of the Notch-1 gene.


EMBO Reports | 2009

RelA/p65 functions to maintain cellular senescence by regulating genomic stability and DNA repair

Jingxin Wang; Naduparambil K. Jacob; Katherine J. Ladner; Amer A. Beg; James D. Perko; Stephan M. Tanner; Sandya Liyanarachchi; Richard Fishel; Denis C. Guttridge

Nuclear factor (NF)‐κB is a positive regulator of tumour development and progression, but how it functions in normal cells leading to oncogenesis is not clear. As cellular senescence has proven to be an intrinsic tumour suppressor mechanism that cells must overcome to establish deregulated growth, we used primary fibroblasts to follow NF‐κB function in cells transitioning from senescence to subsequent immortalization. Our findings show that RelA/p65−/− murine fibroblasts immortalize at considerably faster rates than RelA/p65+/+ cells. The ability of RelA/p65−/− fibroblasts to escape senescence earlier is due to their genomic instability, characterized by high frequencies of DNA mutations, gene deletions and gross chromosomal translocations. This increase in genomic instability is closely related to a compromised DNA repair that occurs in both murine RelA/p65−/− fibroblasts and tissues. Significantly, these results can also be duplicated in human fibroblasts lacking NF‐κB. Altogether, our findings present a fresh perspective on the role of NF‐κB as a tumour suppressor, which acts in pre‐neoplastic cells to maintain cellular senescence by promoting DNA repair and genomic stability.


Science Signaling | 2013

miR-29 acts as a decoy in sarcomas to protect the tumor suppressor A20 mRNA from degradation by HuR.

Mumtaz Yaseen Balkhi; Iwenofu Oh; Bakkar N; Katherine J. Ladner; Dawn S. Chandler; Peter J. Houghton; Cheryl A. London; William G. Kraybill; Danilo Perrotti; Carlo M. Croce; Charles Keller; Denis C. Guttridge

Loss of the decoy function of miR-29 destabilizes A20 transcripts, contributing to oncogenic activation of the transcription factor NF-κB in sarcomas. Decoy MicroRNAs as Tumor Suppressors MicroRNAs are noncoding RNAs that regulate gene expression. In sarcomas, the expression of the microRNA miR-29 is suppressed by NF-κB (nuclear factor κB). Balkhi et al. found that human sarcomas had decreased abundance of miR-29 and A20, an inhibitor of NF-κB, compared with normal tissue or cells but increased abundance of HuR, an RNA binding protein that has binding sites in both miR-29 and the A20 transcript. Inhibition of miR-29 increased HuR-mediated degradation of A20 mRNA, resulting in reduced A20 protein and activation of NF-κB. Overexpression of miR-29 in sarcoma cells inhibited the interaction of A20 mRNA with both HuR and the RNA-induced silencing complex subunit Ago2, increasing the abundance of A20 mRNA and protein. Thus, miR-29 functions as a decoy that prevents HuR-mediated degradation of A20, and loss of this pathway may contribute to NF-κB signaling in sarcoma. In sarcoma, the activity of NF-κB (nuclear factor κB) reduces the abundance of the microRNA (miRNA) miR-29. The tumor suppressor A20 [also known as TNFAIP3 (tumor necrosis factor–α–induced protein 3)] inhibits an upstream activator of NF-κB and is often mutated in lymphomas. In a panel of human sarcoma cell lines, we found that the activation of NF-κB was increased and, although the abundance of A20 protein and mRNA was decreased, the gene encoding A20 was rarely mutated. The 3′ untranslated region (UTR) of A20 mRNA has conserved binding sites for both of the miRNAs miR-29 and miR-125. Whereas the expression of miR-125 was increased in human sarcoma tissue, that of miR-29 was decreased in most samples. Overexpression of miR-125 decreased the abundance of A20 mRNA, whereas reconstituting miR-29 in sarcoma cell lines increased the abundance of A20 mRNA and protein. By interacting directly with the RNA binding protein HuR (human antigen R; also known as ELAVL1), miR-29 prevented HuR from binding to the A20 3′UTR and recruiting the RNA degradation complex RISC (RNA-induced silencing complex), suggesting that miR-29 can act as a decoy for HuR, thus protecting A20 transcripts. Decreased miR-29 and A20 abundance in sarcomas correlated with increased activity of NF-κB and decreased expression of genes associated with differentiation. Together, the findings reveal a unique role of miR-29 and suggest that its absence may contribute to sarcoma tumorigenesis.


Cancer Research | 2008

IFN-alpha and bortezomib overcome Bcl-2 and Mcl-1 overexpression in melanoma cells by stimulating the extrinsic pathway of apoptosis.

Gregory B. Lesinski; Ene T. Raig; Kristan D. Guenterberg; Lloyd Brown; Michael R. Go; Nisha Shah; Adrian Lewis; Megan Quimper; Erinn M. Hade; Gregory S. Young; Abhik Ray Chaudhury; Katherine J. Ladner; Denis C. Guttridge; Page Bouchard; William E. Carson

We hypothesized that IFN-alpha would enhance the apoptotic activity of bortezomib on melanoma cells. Combined treatment with bortezomib and IFN-alpha induced synergistic apoptosis in melanoma and other solid tumor cell lines. Apoptosis was associated with processing of procaspase-3, procaspase-7, procaspase-8, and procaspase-9 and with cleavage of Bid and poly(ADP-ribose) polymerase. Bortezomib plus IFN-alpha was effective at inducing apoptosis in melanoma cells that overexpressed Bcl-2 or Mcl-1, suggesting that this treatment combination can overcome mitochondrial pathways of cell survival and resistance to apoptosis. The proapoptotic effects of this treatment combination were abrogated by a caspase-8 inhibitor, led to increased association of Fas and FADD before the onset of cell death, and were significantly reduced in cells transfected with a dominant-negative FADD construct or small interfering RNA targeting Fas. These data suggest that bortezomib and IFN-alpha act through the extrinsic pathway of apoptosis via FADD-induced caspase-8 activation to initiate cell death. Finally, bortezomib and IFN-alpha displayed statistically significant antitumor activity compared with either agent alone in both the B16 murine model of melanoma and in athymic mice bearing human A375 xenografts. These data support the future clinical development of bortezomib and IFN-alpha for malignant melanoma.


Cancer Cell | 2016

Serine/Threonine Kinase MLK4 Determines Mesenchymal Identity in Glioma Stem Cells in an NF-κB-dependent Manner

Sung Hak Kim; Ravesanker Ezhilarasan; Emma Phillips; Daniel Gallego-Perez; Amanda Sparks; David Taylor; Katherine J. Ladner; Takuya Furuta; Hemragul Sabit; Rishi Raj Chhipa; Ju Hwan Cho; Ahmed Mohyeldin; Samuel Beck; Kazuhiko Kurozumi; Toshihiko Kuroiwa; Ryoichi Iwata; Akio Asai; Jonghwan Kim; Erik P. Sulman; Shi Yuan Cheng; L. James Lee; Mitsutoshi Nakada; Denis C. Guttridge; Biplab Dasgupta; Violaine Goidts; Krishna P. Bhat; Ichiro Nakano

Activation of nuclear factor κB (NF-κB) induces mesenchymal (MES) transdifferentiation and radioresistance in glioma stem cells (GSCs), but molecular mechanisms for NF-κB activation in GSCs are currently unknown. Here, we report that mixed lineage kinase 4 (MLK4) is overexpressed in MES but not proneural (PN) GSCs. Silencing MLK4 suppresses self-renewal, motility, tumorigenesis, and radioresistance of MES GSCs via a loss of the MES signature. MLK4 binds and phosphorylates the NF-κB regulator IKKα, leading to activation of NF-κB signaling in GSCs. MLK4 expression is inversely correlated with patient prognosis in MES, but not PN high-grade gliomas. Collectively, our results uncover MLK4 as an upstream regulator of NF-κB signaling and a potential molecular target for the MES subtype of glioblastomas.


Journal of Cell Biology | 2012

IKKα and alternative NF-κB regulate PGC-1β to promote oxidative muscle metabolism

Nadine Bakkar; Katherine J. Ladner; Benjamin D. Canan; Sandya Liyanarachchi; Naresh C. Bal; Meghna Pant; Muthu Periasamy; Qiutang Li; Paul M. L. Janssen; Denis C. Guttridge

Alternative NF-κB signaling modulates the activity of PGC-1β to promote oxidative metabolism in skeletal muscle.

Collaboration


Dive into the Katherine J. Ladner's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Charles Keller

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Swarnali Acharyya

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Carlo M. Croce

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alfred S.L. Cheng

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar

Brett M. Hall

Nationwide Children's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge