Network


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

Hotspot


Dive into the research topics where William G. Kerr is active.

Publication


Featured researches published by William G. Kerr.


Nature Medicine | 2005

Inhibiting Stat3 signaling in the hematopoietic system elicits multicomponent antitumor immunity

Marcin Kortylewski; Maciej Kujawski; Tianhong Wang; Sheng Wei; Shumin M. Zhang; Shari Pilon-Thomas; Guilian Niu; Heidi Kay; James J. Mulé; William G. Kerr; Richard Jove; Drew M. Pardoll; Hua Yu

The immune system can act as an extrinsic suppressor of tumors. Therefore, tumor progression depends in part on mechanisms that downmodulate intrinsic immune surveillance. Identifying these inhibitory pathways may provide promising targets to enhance antitumor immunity. Here, we show that Stat3 is constitutively activated in diverse tumor-infiltrating immune cells, and ablating Stat3 in hematopoietic cells triggers an intrinsic immune-surveillance system that inhibits tumor growth and metastasis. We observed a markedly enhanced function of dendritic cells, T cells, natural killer (NK) cells and neutrophils in tumor-bearing mice with Stat3−/− hematopoietic cells, and showed that tumor regression requires immune cells. Targeting Stat3 with a small-molecule drug induces T cell– and NK cell–dependent growth inhibition of established tumors otherwise resistant to direct killing by the inhibitor. Our findings show that Stat3 signaling restrains natural tumor immune surveillance and that inhibiting hematopoietic Stat3 in tumor-bearing hosts elicits multicomponent therapeutic antitumor immunity.


Immunity | 2003

A Critical Role for Stat3 Signaling in Immune Tolerance

Fengdong Cheng; Hongwei Wang; Alex G. Cuenca; Mei Huang; Tomar Ghansah; Jason Brayer; William G. Kerr; Kiyoshi Takeda; Shizuo Akira; Stephen P. Schoenberger; Hua Yu; Richard Jove; Eduardo M. Sotomayor

Antigen-presenting cells (APCs) can induce T cell activation as well as T cell tolerance. The molecular mechanisms by which APCs regulate this critical decision of the immune system are not well understood. Here we show that Stat3 signaling plays a critical role in the induction of antigen-specific T cell tolerance. Targeted disruption of Stat3 signaling in APCs resulted in priming of antigen-specific CD4(+) T cells in response to an otherwise tolerogenic stimulus in vivo. Furthermore, APCs devoid of Stat3 effectively break antigen-specific T cell anergy in vitro. Conversely, increased Stat3 activity in APCs led to impaired antigen-specific T cell responses. Stat3 signaling provides, therefore, a novel molecular target for manipulation of immune activation/tolerance, a central decision with profound implications in autoimmunity, transplantation, and cancer immunotherapy.


Science Signaling | 2010

TREM2- and DAP12-Dependent Activation of PI3K Requires DAP10 and Is Inhibited by SHIP1

Qisheng Peng; Shikha Malhotra; James Torchia; William G. Kerr; Coggeshall Km; Mary Beth Humphrey

The inositol phosphatase SHIP1 binds to a receptor-adaptor complex on osteoclasts to prevent recruitment of PI3K and inhibit receptor signaling. Keeping Osteoclasts in Check Bone resorption is mediated by cells known as osteoclasts, which develop from myeloid precursor cells. Differentiation and activation of osteoclasts depend on the signaling of receptors whose responses are mediated by the adaptor protein DAP12. One such receptor is TREM2, which drives osteoclastogenesis; deficiencies in either DAP12 or TREM2 result in similar phenotypes. TREM2- and DAP12-dependent signaling also inhibits some Toll-like receptor (TLR)–dependent responses in macrophages, another myeloid cell type. Thus, the TREM2-DAP12 signaling axis may be both stimulatory and inhibitory; however, little is known about how DAP12 functions in these different contexts. Peng et al. found that ligation of TREM2 with a cross-linking antibody triggered DAP12-dependent osteoclastogenesis that required the recruitment of phosphatidylinositol 3-kinase (PI3K) to the TREM2-DAP12 signaling complex. This signaling pathway was inhibited by the Src homology 2 (SH2) domain–dependent recruitment to TREM2-DAP12 of the inositol phosphatase SHIP1, which prevented PI3K from binding to DAP12. In addition, signaling downstream of other receptors on osteoclasts differentially recruited SHIP1 to DAP12. The authors thus propose that the stimulatory or inhibitory nature of TREM2-DAP12 signaling is regulated by SHIP1. The activation and fusion of macrophages and of osteoclasts require the adaptor molecule DNAX-activating protein of 12 kD (DAP12), which contains immunoreceptor tyrosine-based activation motifs (ITAMs). TREM2 (triggering receptor expressed on myeloid cells–2) is the main DAP12-associated receptor in osteoclasts and, similar to DAP12 deficiency, loss of TREM2 in humans leads to Nasu-Hakola disease, which is characterized by bone cysts and dementia. Furthermore, in vitro experiments have shown that deficiency in DAP12 or TREM2 leads to impaired osteoclast development and the formation of mononuclear osteoclasts. Here, we demonstrate that the ligation of TREM2 activated phosphatidylinositol 3-kinase (PI3K), extracellular signal–regulated kinase 1 (ERK1) and ERK2, and the guanine nucleotide exchange factor Vav3; induced the mobilization of intracellular calcium (Ca2+) and the reorganization of actin; and prevented apoptosis. The signaling adaptor molecule DAP10 played a key role in the TREM2- and DAP12-dependent recruitment of PI3K to the signaling complex. Src homology 2 (SH2) domain–containing inositol phosphatase-1 (SHIP1) inhibited TREM2- and DAP12-induced signaling by binding to DAP12 in an SH2 domain–dependent manner and preventing the recruitment of PI3K to DAP12. These results demonstrate a previously uncharacterized interaction of SHIP1 with DAP12 that functionally limits TREM2- and DAP12-dependent signaling and identify a mechanism through which SHIP1 regulates key ITAM-containing receptors by directly blocking the binding and activation of PI3K.


Journal of Immunology | 2004

Expansion of Myeloid Suppressor Cells in SHIP-Deficient Mice Represses Allogeneic T Cell Responses

Tomar Ghansah; Kim H. T. Paraiso; Steven Highfill; Caroline Desponts; Sarah May; Joseph K. McIntosh; Jia-Wang Wang; John M. Ninos; Jason Brayer; Fengdong Cheng; Eduardo M. Sotomayor; William G. Kerr

Previously we demonstrated that SHIP−/− mice accept allogeneic bone marrow transplants (BMT) without significant acute graft-vs-host disease (GvHD). In this study we show that SHIP−/− splenocytes and lymph node cells are poor stimulators of allogeneic T cell responses that cause GvHD. Intriguingly, SHIP−/− splenocytes prime naive T cell responses to peptide epitopes, but, conversely, are partially impaired for priming T cell responses to whole Ag. However, dendritic cells (DC) purified from SHIP−/− splenocytes prime T cell responses to allogeneic targets, peptide epitopes, and whole Ag as effectively as SHIP+/+ DC. These findings point to an extrinsic effect on SHIP−/− DC that impairs priming of allogeneic T cell responses. Consistent with this extrinsic effect, we found that a dramatic expansion of myeloid suppressor cells in SHIP−/− mice impairs priming of allogeneic T cells. These findings suggest that SHIP expression or its activity could be targeted to selectively compromise T cell responses that mediate GvHD and graft rejection.


Journal of Immunology | 2001

Identification of a Novel Lipopolysaccharide-Inducible Gene with Key Features of Both a Kinase Anchor Proteins and chs1/beige Proteins

Jia-Wang Wang; Julie M. Howson; Edward M Haller; William G. Kerr

Mutations in chs1/beige result in a deficiency in intracellular transport of vesicles that leads to a generalized immunodeficiency in mice and humans. The function of NK cells, CTL, and granulocytes is impaired by these mutations, indicating that polarized trafficking of vesicles is controlled by CHS1/beige proteins. However, a molecular explanation for this defect has not been identified. Here we describe a novel gene with orthologues in mice, humans, and flies that contains key features of both chs1/beige and A kinase anchor genes. We designate this novel gene lba for LPS-responsive, beige-like anchor gene. Expression of lba is induced after LPS stimulation of B cells and macrophages. In addition, lba is expressed in many other tissues in the body and has three distinct mRNA isoforms that are differentially expressed in various tissues. Strikingly, LBA-green-fluorescent protein (GFP) fusion proteins are localized to vesicles after LPS stimulation. Confocal microscopy indicates this protein is colocalized with the trans-Golgi complex and some lysosomes. Further analysis by immunoelectron microscopy demonstrates that LBA-GFP fusion protein can localize to endoplasmic reticulum, plasma membrane, and endocytosis vesicles in addition to the trans-Golgi complex and lysosomes. We hypothesize that LBA/CHS1/BG proteins function in polarized vesicle trafficking by guiding intracellular vesicles to activated receptor complexes and thus facilitate polarized secretion and/or membrane deposition of immune effector molecules.


Journal of Immunology | 2010

SHIP1 inhibition increases immunoregulatory capacity and triggers apoptosis of hematopoietic cancer cells.

Robert Brooks; Gwenny M. Fuhler; Sonia Iyer; Michelle J. Smith; Mi-Young Park; Kim H. T. Paraiso; Robert W. Engelman; William G. Kerr

Genetic studies revealed that SHIP1 limits blood cell production and immune regulatory cell numbers in vivo. We postulated that molecular targeting of SHIP1 might enhance blood cell production and increase immunoregulatory capacity. In this study, we report the identification of a chemical inhibitor of SHIP1, 3 α-aminocholestane (3AC). Treatment with 3AC significantly expands the myeloid immunoregulatory cell compartment and impairs the ability of peripheral lymphoid tissues to prime allogeneic T cell responses. In addition, 3AC treatment profoundly increases granulocyte production without triggering the myeloid-associated lung consolidation observed in SHIP1−/− mice. Moreover, 3AC also enhances RBC, neutrophil, and platelet recovery in myelosuppressed hosts. Intriguingly, we also find that chemical inhibition of SHIP1 triggers apoptosis of blood cancer cells. Thus, SHIP1 inhibitors represent a novel class of small molecules that have the potential to enhance allogeneic transplantation, boost blood cell production, and improve the treatment of hematologic malignancies.


Annals of the New York Academy of Sciences | 2011

Inhibitor and activator: dual functions for SHIP in immunity and cancer.

William G. Kerr

SHIP1 is at the nexus of intracellular signaling pathways in immune cells that mediate bone marrow (BM) graft rejection, production of inflammatory and immunosuppressive cytokines, immunoregulatory cell formation, the BM niche that supports development of the immune system, and immune cancers. This review summarizes how SHIP participates in normal immune physiology or the pathologies that result when SHIP is mutated. This review also proposes that SHIP can have either inhibitory or activating roles in cell signaling that are determined by whether signaling pathways distal to PI3K are promoted by SHIPs substrate (PI(3,4,5)P3) or its product (PI(3,4)P2). This review also proposes the “two PIP hypothesis” that postulates that both SHIPs product and its substrate are necessary for a cancer cell to achieve and sustain a malignant state. Finally, due to the recent discovery of small molecule antagonists and agonists for SHIP, this review discusses potential therapeutic settings where chemical modulation of SHIP might be of benefit.


Journal of Immunology | 2007

Induced SHIP deficiency expands myeloid regulatory cells and abrogates graft-versus-host disease

Kim H. T. Paraiso; Tomar Ghansah; Amy L. Costello; Robert W. Engelman; William G. Kerr

Graft-vs-host disease (GVHD) is the leading cause of treatment-related death in allogeneic bone marrow (BM) transplantation. Immunosuppressive strategies to control GVHD are only partially effective and often lead to life-threatening infections. We previously showed that engraftment of MHC-mismatched BM is enhanced and GVHD abrogated in recipients homozygous for a germline SHIP mutation. In this study, we report the development of a genetic model in which SHIP deficiency can be induced in adult mice. Using this model, we show that the induction of SHIP deficiency in adult mice leads to a rapid and significant expansion of myeloid suppressor cells in peripheral lymphoid tissues. Consistent with expansion of myeloid suppressor cells, splenocytes and lymph node cells from adult mice with induced SHIP deficiency are significantly compromised in their ability to prime allogeneic T cell responses. These results demonstrate that SHIP regulates homeostatic signals for these immunoregulatory cells in adult physiology. Consistent with these findings, induction of SHIP deficiency before receiving a T cell-replete BM graft abrogates acute GVHD. These findings indicate strategies that target SHIP could increase the efficacy and utility of allogeneic BM transplantation, and thereby provide a curative therapy for a wide spectrum of human diseases.


Journal of Immunology | 2007

Inappropriate Recruitment and Activity by the Src Homology Region 2 Domain-Containing Phosphatase 1 (SHP1) Is Responsible for Receptor Dominance in the SHIP-Deficient NK Cell

Joseph Wahle; Kim H. T. Paraiso; Robert D. Kendig; Harshani R. Lawrence; Liwei Chen; Jerry Wu; William G. Kerr

We have previously demonstrated that the NKR repertoire is profoundly disrupted by SHIP deficiency. This repertoire disruption is characterized by receptor dominance where inhibitory signals from 2B4 repress killing of complex targets expressing MHC class I and activating ligands. In this study, we examine the molecular basis of receptor dominance in SHIP−/− NK cells. In this study, we show that in SHIP−/− NK cells there is a pronounced bias toward the 2B4 long isoform. We have also characterized signaling molecules recruited to 2B4 in SHIP−/− NK cells. Interestingly, we find that ∼10- to 16-fold more Src homology region 2 domain-containing phosphatase 1 (SHP1) is recruited to 2B4 in SHIP−/− NK cells when compared with wild type. Consistent with SHP1 overrecruitment, treatment with sodium orthovanadate or a novel inhibitor with micromolar activity against SHP1 restores the ability of SHIP−/− NK cells to kill Rae1+ RMA and M157+ targets. These findings define the molecular basis for hyporesponsiveness by SHIP-deficient NK cells.


Molecular Medicine | 2012

Therapeutic Potential of SH2 Domain-Containing Inositol-5′-Phosphatase 1 (SHIP1) and SHIP2 Inhibition in Cancer

Gwenny M. Fuhler; Robert Brooks; Bonnie B. Toms; Sonia Iyer; Elizabeth A. Gengo; Mi Young Park; Matthew Gumbleton; Dennis R. Viernes; John D. Chisholm; William G. Kerr

Many tumors present with increased activation of the phosphatidylinositol 3-kinase (PI3K)-PtdIns(3,4,5)P3-protein kinase B (PKB/Akt) signaling pathway. It has long been thought that the lipid phosphatases SH2 domain-containing inositol-5′-phosphatase 1 (SHIP1) and SHIP2 act as tumor suppressors by counteracting with the survival signal induced by this pathway through hydrolysis or PtdIns(3/4/5)P3 to PtdIns(3,4)P2. However, a growing body of evidence suggests that PtdInd(3,4)P2 is capable of, and essential for, Akt activation, thus suggesting a potential role for SHIP1/2 enzymes as proto-oncogenes. We recently described a novel SHIP1-selective chemical inhibitor (3α-aminocholestane (3AC)) that is capable of killing malignant hematologic cells. In this study, we further investigate the biochemical consequences of 3AC treatment in multiple myeloma (MM) and demonstrate that SHIP1 inhibition arrests MM cell lines in either G0/G1 or G2/M stages of the cell cycle, leading to caspase activation and apoptosis. In addition, we show that in vivo growth of MM cells is blocked by treatment of mice with the SHIP1 inhibitor 3AC. Furthermore, we identify three novel pan-SHIP1/2 inhibitors that efficiently kill MM cells through G2/M arrest, caspase activation and apoptosis induction. Interestingly, in SHIP2-expressing breast cancer cells that lack SHIP1 expression, pan-SHIP1/2 inhibition also reduces viable cell numbers, which can be rescued by addition of exogenous PtdIns(3,4)P2. In conclusion, this study shows that inhibition of SHIP1 and SHIP2 may have broad clinical application in the treatment of multiple tumor types.

Collaboration


Dive into the William G. Kerr's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robert W. Engelman

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Caroline Desponts

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Kim H. T. Paraiso

State University of New York Upstate Medical University

View shared research outputs
Top Co-Authors

Avatar

Sandra Fernandes

State University of New York Upstate Medical University

View shared research outputs
Top Co-Authors

Avatar

Jia-Wang Wang

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

John M. Ninos

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Sonia Iyer

State University of New York Upstate Medical University

View shared research outputs
Top Co-Authors

Avatar

Matthew Gumbleton

State University of New York Upstate Medical University

View shared research outputs
Top Co-Authors

Avatar

Tomar Ghansah

University of South Florida

View shared research outputs
Researchain Logo
Decentralizing Knowledge