Network


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

Hotspot


Dive into the research topics where Carol Clayberger is active.

Publication


Featured researches published by Carol Clayberger.


Tissue Antigens | 2009

Biology and clinical relevance of granulysin.

Alan M. Krensky; Carol Clayberger

Granulysin is a cytolytic and proinflammatory molecule first identified by a screen for genes expressed late (3-5 days) after activation of human peripheral blood mononuclear cells. Granulysin is present in cytolytic granules of cytotoxic T lymphocytes and natural killer cells. Granulysin is made in a 15-kDa form that is cleaved into a 9-kDa form at both the amino and the carboxy termini. The 15-kDa form is constitutively secreted, and its function remains poorly understood. The 9-kDa form is released by receptor-mediated granule exocytosis. Nine kiloDalton granulysin is broadly cytolytic against tumors and microbes, including gram-positive and gram-negative bacteria, fungi/yeast and parasites. It kills the causative agents of both tuberculosis and malaria. Granulysin is also a chemoattractant for T lymphocytes, monocytes and other inflammatory cells and activates the expression of a number of cytokines, including regulated upon activation T cell expressed and secreted (RANTES), monocyte chemoattractant protein (MCP)-1, MCP-3, macrophage inflammatory protein (MIP)-1 alpha, interleukin (IL)-10, IL-1, IL-6 and interferon (IFN)-alpha. Granulysin is implicated in a myriad of diseases including infection, cancer, transplantation, autoimmunity, skin and reproductive maladies. Small synthetic forms of granulysin are being developed as novel antibiotics. Studies of the full-length forms may give rise to new diagnostics and therapeutics for use in a wide variety of diseases.


Cancer Immunology, Immunotherapy | 2011

Monocyte-derived DC maturation strategies and related pathways: a transcriptional view

Luciano Castiello; Marianna Sabatino; Ping Jin; Carol Clayberger; Francesco M. Marincola; Alan M. Krensky; David F. Stroncek

Ex vivo production of highly stimulator mature dendritic cells (DCs) for cellular therapy has been used to treat different pathological conditions with the aim of inducing a specific immune response. In the last decade, several protocols have been developed to mature monocyte-derived DCs: each one has led to the generation of DCs showing different phenotypes and stimulatory abilities, but it is not yet known which one is the best for inducing effective immune responses. We grouped several different maturation protocols according to the downstream pathways they activated and reviewed the shared features at a transcriptomic level to reveal the potential of DCs matured by each protocol to develop Th-polarized immune responses.


Cell | 2014

Cytotoxic Cells Kill Intracellular Bacteria through Granulysin-Mediated Delivery of Granzymes

Michael Walch; Farokh Dotiwala; Sachin Mulik; Jerome Thiery; Tomas Kirchhausen; Carol Clayberger; Alan M. Krensky; Denis Martinvalet; Judy Lieberman

When killer lymphocytes recognize infected cells, perforin delivers cytotoxic proteases (granzymes) into the target cell to trigger apoptosis. What happens to intracellular bacteria during this process is unclear. Human, but not rodent, cytotoxic granules also contain granulysin, an antimicrobial peptide. Here, we show that granulysin delivers granzymes into bacteria to kill diverse bacterial strains. In Escherichia coli, granzymes cleave electron transport chain complex I and oxidative stress defense proteins, generating reactive oxygen species (ROS) that rapidly kill bacteria. ROS scavengers and bacterial antioxidant protein overexpression inhibit bacterial death. Bacteria overexpressing a GzmB-uncleavable mutant of the complex I subunit nuoF or strains that lack complex I still die, but more slowly, suggesting that granzymes disrupt multiple vital bacterial pathways. Mice expressing transgenic granulysin are better able to clear Listeria monocytogenes. Thus killer cells play an unexpected role in bacterial defense.


Journal of Experimental Medicine | 2011

KLF13 sustains thymic memory-like CD8+ T cells in BALB/c mice by regulating IL-4–generating invariant natural killer T cells

Dazhi Lai; Tianhong Wang; Jane Hu-Li; Masaki Terabe; Jay A. Berzofsky; Carol Clayberger; Alan M. Krensky

Transcription factor KLF13 regulates the elevated numbers of iNKT cells in the BALB/c versus C57BL/6 thymus that results in production of sufficient levels of IL-4 to generate memory-like CD8+ T cells.


Journal of Immunology | 2011

Granulysin Delivered by Cytotoxic Cells Damages Endoplasmic Reticulum and Activates Caspase-7 in Target Cells

Reena V. Saini; Christine Wilson; Michael W. Finn; Tianhong Wang; Alan M. Krensky; Carol Clayberger

Granulysin is a human cytolytic molecule present in cytotoxic granules with perforin and granzymes. Recombinant 9-kDa granulysin kills a variety of microbes, including bacteria, yeast, fungi, and parasites, and induces apoptosis in tumor cells by causing intracellular calcium overload, mitochondrial damage, and activation of downstream caspases. Reasoning that granulysin delivered by cytotoxic cells may work in concert with other molecules, we crossed granulysin transgenic (GNLY+/−) mice onto perforin (perf)- or granzyme B (gzmb)-deficient mice to examine granulysin-mediated killing in a more physiologic whole-cell system. Splenocytes from these animals were activated in vitro with IL-15 to generate cytolytic T cells and NK cells. Cytotoxic cells expressing granulysin require perforin, but not granzyme B, to cause apoptosis of targets. Whereas granzyme B induces mitochondrial damage and activates caspases-3 and -9 in targets, cytotoxic cell-delivered granulysin induces endoplasmic reticulum stress and activates caspase-7 with no effect on mitochondria or caspases-3 and -9. In addition, recombinant granulysin and cell-delivered granulysin activate distinct apoptotic pathways in target cells. These findings suggest that cytotoxic cells have evolved multiple nonredundant cell death pathways, enabling host defense to counteract escape mechanisms employed by pathogens or tumor cells.


Journal of Immunology | 2012

15 kDa Granulysin Causes Differentiation of Monocytes to Dendritic Cells but Lacks Cytotoxic Activity

Carol Clayberger; Michael W. Finn; Tianhong Wang; Reena V. Saini; Christine Wilson; Valarie A. Barr; Marianna Sabatino; Luciano Castiello; David F. Stroncek; Alan M. Krensky

Granulysin is expressed as two isoforms by human cytotoxic cells: a single mRNA gives rise to 15 kDa granulysin, a portion of which is cleaved to a 9 kDa protein. Studies with recombinant 9 kDa granulysin have demonstrated its cytolytic and proinflammatory properties, but much less is known about the biologic function of the 15 kDa isoform. In this study, we show that the subcellular localization and functions of 9 and 15 kDa granulysin are largely distinct. Nine kilodalton granulysin is confined to cytolytic granules that are directionally released following target cell recognition. In contrast, 15 kDa granulysin is located in distinct granules that lack perforin and granzyme B and that are released by activated cytolytic cells. Although recombinant 9 kDa granulysin is cytolytic against a variety of tumors and microbes, recombinant 15 kDa granulysin is not. The 15 kDa isoform is a potent inducer of monocytic differentiation to dendritic cells, but the 9 kDa isoform is not. In vivo, mice expressing granulysin show markedly improved antitumor responses, with increased numbers of activated dendritic cells and cytokine-producing T cells. Thus, the distinct functions of granulysin isoforms have major implications for diagnosis and potential new therapies for human disease.


Current Opinion in Organ Transplantation | 2009

Cytolytic molecules in rejection.

Carol Clayberger

Purpose of reviewAcute and chronic rejection are major problems in clinical transplantation. Rejection is largely mediated by natural killer (NK) and T cells that use cytolytic molecules, including perforin, granzymes, granulysin, and Fas ligand, to eliminate the allograft. The purpose of this review is to inform the reader of recent advances in our understanding of the roles of cytolytic molecules in rejection and their potential as biomarkers of rejection. Recent findingsAlthough it is well accepted that T cells are the major effector cells in acute rejection, there is an increasing recognition that cells of the innate immune system, and in particular NK cells, also play a major role in allograft rejection. SummaryBoth NK cells and cytotoxic T cells contribute to acute rejection. The major molecules involved include perforin, granzymes, granulysin, and Fas ligand. Molecular profiles that include these and other molecules may allow better management of organ allograft recipients.


Biology of Reproduction | 2012

The Reproductive Phenotype of Mice Null for Transcription Factor Krüppel-Like Factor 13 Suggests Compensatory Function of Family Member Krüppel-Like Factor 9 in the Peri-Implantation Uterus

Melissa E. Heard; John Mark P. Pabona; Carol Clayberger; Alan M. Krensky; Frank A. Simmen; Rosalia C. M. Simmen

ABSTRACT The ovarian hormones estrogen and progesterone promote uterine receptivity and successful pregnancy through their cognate receptors functioning in concert with context-dependent nuclear coregulators. Previously, we showed that the transcription factor Krüppel-like factor (KLF) 9 is a progesterone receptor (PGR) coactivator in the uterus and that mice null for Klf9 exhibit subfertility and reduced progesterone sensitivity. The highly related family member KLF13 displays increased expression in uteri of pregnant and nonpregnant Klf9 null mice and similarly regulates PGR-mediated transactivation in endometrial stromal cells. However, a uterine phenotype with loss of Klf13 has not been reported. In the present study, we demonstrate that Klf13 deficiency in mice did not compromise female fertility and pregnancy outcome. Klf13 null females had litter sizes, numbers of implanting embryos, uterine morphology, and ovarian steroid hormone production comparable to those of wild-type (WT) counterparts. Further, pregnant WT and Klf13 null females at Day Postcoitum (DPC) 3.5 had similar uterine Pgr, estrogen receptor, and Wnt-signaling component transcript levels. Nuclear levels of KLF9 were higher in Klf13 null than in WT uteri at DPC 3.5, albeit whole-tissue KLF9 protein and transcript levels did not differ between genotypes. The lack of a similar induction of nuclear KLF9 levels in uteri of virgin Klf13(−/−) mice relative to WT uteri was associated with lower stromal PGR expression. In differentiating human endometrial stromal cells, coincident KLF9/KLF13 knockdown by small interfering RNA targeting reduced decidualization-associated PRL expression, whereas KLF9 and KLF13 knockdowns alone reduced transcript levels of WNT4 and BMP2, respectively. Results suggest that KLF9 and KLF13 functionally compensate in peri-implantation uterus for pregnancy success.


Journal of Immunology | 2014

KLF13 Cooperates with c-Maf To Regulate IL-4 Expression in CD4+ T Cells

Seok Joo Kwon; Juan Crespo-Barreto; Wei Zhang; Tianhong Wang; Dong Seok Kim; Alan M. Krensky; Carol Clayberger

Kruppel-like factor (KLF) 13 is a transcription factor that positively regulates expression of the chemokine RANTES 3–5 d after activation of T cells. In this study, we document a key role for KLF13 in the expression of IL-4 in CD4+ T cells. Gene expression analysis in activated T cells from Klf13−/− mice showed that IL-4, along with other Th2 cytokine genes, was downregulated when compared with cells from wild-type mice. The decreased levels of IL-4 were not associated with changes in expression of the Th2-inducing transcription factors GATA3 or c-Maf. Additional analysis revealed that KLF13 directly binds to IL-4 promoter regions and synergizes with c-Maf to positively regulate IL-4 expression. These results indicate that KLF13 is a positive regulator for differentiation of Th2 cells, as part of the transcriptional machinery that regulates IL-4 production in Th2 cells.


Protein Expression and Purification | 2011

Expression and purification of 15 kDa granulysin utilizing an insect cell secretion system.

Michael W. Finn; Carol Clayberger; Alan M. Krensky

Granulysin is an antimicrobial and proinflammatory protein expressed in activated human T cells and natural killer cells. A single mRNA produces the 15 kDa isoform which is then cleaved at the amino and carboxy termini to produce the 9 kDa isoform. Recombinant 9 kDa granulysin has been studied in detail but little is known about the function of the 15 kDa isoform, and no protocol has been published describing expression and purification of this form. Two commercially available preparations of the recombinant 15 kDa granulysin contain tags that may affect function. Here we describe for the first time a method to produce 15 kDa granulysin as a secreted protein from insect cells. The 15 kDa granulysin is purified using a HiTrap Heparin column and a Resource S column. A typical a yield of purified 15 kDa granulysin is 0.6 mg/L of insect cell supernatant.

Collaboration


Dive into the Carol Clayberger's collaboration.

Top Co-Authors

Avatar

Alan M. Krensky

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Michael W. Finn

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Tianhong Wang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Archana Roy

Northwestern University

View shared research outputs
Top Co-Authors

Avatar

David F. Stroncek

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Luciano Castiello

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Marianna Sabatino

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge