Network


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

Hotspot


Dive into the research topics where Jeff S. Pawlikowski is active.

Publication


Featured researches published by Jeff S. Pawlikowski.


Journal of Cell Biology | 2013

Lysosome-mediated processing of chromatin in senescence

Andre Ivanov; Jeff S. Pawlikowski; Indrani Manoharan; John van Tuyn; David M. Nelson; Taranjit Singh Rai; Parisha P. Shah; Graeme Hewitt; Viktor I. Korolchuk; João F. Passos; Hong Wu; Shelley L. Berger; Peter D. Adams

Senescent cells extrude fragments of chromatin from the nucleus into the cytoplasm, where they are processed by an autophagic/lysosomal pathway.


The Journal of Pathology | 2012

Activation of Wnt signalling promotes development of dysplasia in Barrett's oesophagus

Lisa H Moyes; Hamish McEwan; Sorina Radulescu; Jeff S. Pawlikowski; Catherine G. Lamm; Colin Nixon; Owen J. Sansom; James J. Going; Grant Fullarton; Peter D. Adams

Barretts oesophagus is a precursor of oesophageal adenocarcinoma, via intestinal metaplasia and dysplasia. Risk of cancer increases substantially with dysplasia, particularly high‐grade dysplasia. Thus, there is a clinical need to identify and treat patients with early‐stage disease (metaplasia and low‐grade dysplasia) that are at high risk of cancer. Activated Wnt signalling is critical for normal intestinal development and homeostasis, but less so for oesophageal development. Therefore, we asked whether abnormally increased Wnt signalling contributes to the development of Barretts oesophagus (intestinal metaplasia) and/or dysplasia. Forty patients with Barretts metaplasia, dysplasia or adenocarcinoma underwent endoscopy and biopsy. Mice with tamoxifen‐ and β‐naphthoflavone‐induced expression of activated β‐catenin were used to up‐regulate Wnt signalling in mouse oesophagus. Immunohistochemistry of β‐catenin, Ki67, a panel of Wnt target genes, and markers of intestinal metaplasia was performed on human and mouse tissues. In human tissues, expression of nuclear activated β‐catenin was found in dysplasia, particularly high grade. Barretts metaplasia did not show high levels of activated β‐catenin. Up‐regulation of Ki67 and Wnt target genes was also mostly associated with high‐grade dysplasia. Aberrant activation of Wnt signalling in mouse oesophagus caused marked tissue disorganization with features of dysplasia, but only selected molecular indicators of metaplasia. Based on these results in human tissues and a mouse model, we conclude that abnormal activation of Wnt signalling likely plays only a minor role in initiation of Barretts metaplasia but a more critical role in progression to dysplasia. Copyright


Cancer Research | 2015

Mdm2 and Aurora Kinase A Inhibitors Synergize to Block Melanoma Growth by Driving Apoptosis and Immune Clearance of Tumor Cells

Anna E. Vilgelm; Jeff S. Pawlikowski; Yan Liu; Oriana E. Hawkins; Tyler A. Davis; Jessica Smith; Kevin P. Weller; Linda W. Horton; Colt M. McClain; Gregory D. Ayers; David C. Turner; David C. Essaka; Clinton F. Stewart; Jeffrey A. Sosman; Mark C. Kelley; Jeffrey Ecsedy; Jeffrey N. Johnston; Ann Richmond

Therapeutics that induce cancer cell senescence can block cell proliferation and promote immune rejection. However, the risk of tumor relapse due to senescence escape may remain high due to the long lifespan of senescent cells that are not cleared. Here, we show how combining a senescence-inducing inhibitor of the mitotic kinase Aurora A (AURKA) with an MDM2 antagonist activates p53 in senescent tumors harboring wild-type 53. In the model studied, this effect is accompanied by proliferation arrest, mitochondrial depolarization, apoptosis, and immune clearance of cancer cells by antitumor leukocytes in a manner reliant upon Ccl5, Ccl1, and Cxcl9. The AURKA/MDM2 combination therapy shows adequate bioavailability and low toxicity to the host. Moreover, the prominent response of patient-derived melanoma tumors to coadministered MDM2 and AURKA inhibitors offers a sound rationale for clinical evaluation. Taken together, our work provides a preclinical proof of concept for a combination treatment that leverages both senescence and immune surveillance to therapeutic ends.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Wnt signaling potentiates nevogenesis

Jeff S. Pawlikowski; Tony McBryan; J. van Tuyn; Mark E. Drotar; Rachael N. Hewitt; Andrea B. Maier; Ayala King; Karen Blyth; Hong Wu; Peter D. Adams

Significance Human benign nevi (moles) are clonal neoplasms that rarely progress to melanoma because their cells (melanocytes) are arrested in a viable but nonproliferating state (senescence). However, at low frequency, nevus melanocytes do progress to melanoma. Consequently, it is important to understand the factors that determine nevus formation and progression to melanoma. We present evidence that repression of a proliferation-promoting cell signaling pathway (Wnt signaling pathway) contributes to senescence of melanocytes in vitro. However, Wnt signaling remains active in some senescent human melanocytes in nevi, and activation of Wnt signaling leads to a delay in melanocyte senescence in a mouse model. We suggest that activated Wnt signaling in human nevi delays senescence to promote nevus formation, and thereafter, persistent Wnt signaling might undermine senescence-mediated tumor suppression. Cellular senescence is a stable proliferation arrest associated with an altered secretory pathway (senescence-associated secretory phenotype). Cellular senescence is also a tumor suppressor mechanism, to which both proliferation arrest and senescence-associated secretory phenotype are thought to contribute. The melanocytes within benign human nevi are a paradigm for tumor-suppressive senescent cells in a premalignant neoplasm. Here a comparison of proliferating and senescent melanocytes and melanoma cell lines by RNA sequencing emphasizes the importance of senescence-associated proliferation arrest in suppression of transformation. Previous studies showed that activation of the Wnt signaling pathway can delay or bypass senescence. Consistent with this, we present evidence that repression of Wnt signaling contributes to melanocyte senescence in vitro. Surprisingly, Wnt signaling is active in many senescent human melanocytes in nevi, and this is linked to histological indicators of higher proliferative and malignant potential. In a mouse, activated Wnt signaling delays senescence-associated proliferation arrest to expand the population of senescent oncogene-expressing melanocytes. These results suggest that Wnt signaling can potentiate nevogenesis in vivo by delaying senescence. Further, we suggest that activated Wnt signaling in human nevi undermines senescence-mediated tumor suppression and enhances the probability of malignancy.


Journal of Cell Science | 2013

Senescence at a glance

Jeff S. Pawlikowski; Peter D. Adams; David M. Nelson

Summary Cellular senescence is a stable proliferation arrest that is associated with extensive cellular remodelling and an altered secretory pathway. Through its numerous inducers that lead to altered gene expression, senescence is able to influence many contrasting functions and pathologies, namely tumour suppression, tumour promotion, wound healing and ageing. As senescence is able to control such important tissue functions, it is now being pinpointed as a possible route for novel therapies. This article and accompanying poster aim to provide a summary of the initiators, pathways and roles of senescence, as well as present examples of senescence and a possible use for senescence in therapy.


PLOS ONE | 2013

The Specificity and Patterns of Staining in Human Cells and Tissues of p16INK4a Antibodies Demonstrate Variant Antigen Binding

Magdalena Sawicka; Jeff S. Pawlikowski; Stephen W. Wilson; Dudley Ferdinando; Hong Wu; Peter D. Adams; David A. Gunn; William L. Parish

The validity of the identification and classification of human cancer using antibodies to detect biomarker proteins depends upon antibody specificity. Antibodies that bind to the tumour-suppressor protein p16INK4a are widely used for cancer diagnosis and research. In this study we examined the specificity of four commercially available anti-p16INK4a antibodies in four immunological applications. The antibodies H-156 and JC8 detected the same 16 kDa protein in western blot and immunoprecipitation tests, whereas the antibody F-12 did not detect any protein in western blot analysis or capture a protein that could be recognised by the H-156 antibody. In immunocytochemistry tests, the antibodies JC8 and H-156 detected a predominately cytoplasmic localised antigen, whose signal was depleted in p16INK4a siRNA experiments. F-12, in contrast, detected a predominately nuclear located antigen and there was no noticeable reduction in this signal after siRNA knockdown. Furthermore in immunohistochemistry tests, F-12 generated a different pattern of staining compared to the JC8 and E6H4 antibodies. These results demonstrate that three out of four commercially available p16INK4a antibodies are specific to, and indicate a mainly cytoplasmic localisation for, the p16INK4a protein. The F-12 antibody, which has been widely used in previous studies, gave different results to the other antibodies and did not demonstrate specificity to human p16INK4a. This work emphasizes the importance of the validation of commercial antibodies, aside to the previously reported use, for the full verification of immunoreaction specificity.


Journal of the National Cancer Institute | 2016

Connecting the Dots: Therapy-Induced Senescence and a Tumor-Suppressive Immune Microenvironment

Anna E. Vilgelm; C. Andrew Johnson; Nripesh Prasad; Jinming Yang; Sheau-Chiann Chen; Gregory D. Ayers; Jeff S. Pawlikowski; Dayanidhi Raman; Jeffrey A. Sosman; Mark C. Kelley; Jeffrey Ecsedy; Yu Shyr; Shawn Levy; Ann Richmond

BACKGROUND Tumor cell senescence is a common outcome of anticancer therapy. Here we investigated how therapy-induced senescence (TIS) affects tumor-infiltrating leukocytes (TILs) and the efficacy of immunotherapy in melanoma. METHODS Tumor senescence was induced by AURKA or CDK4/6 inhibitors (AURKAi, CDK4/6i). Transcriptomes of six mouse tumors with differential response to AURKAi were analyzed by RNA sequencing, and TILs were characterized by flow cytometry. Chemokine RNA and protein expression were determined by quantitative real-time polymerase chain reaction and enzyme-linked immunosorbent assay. Therapeutic response was queried in immunodeficient mice, in mice with CCL5-deficient tumors, and in mice cotreated with CD137 agonist to activate TILs. CCL5 expression in reference to TIS and markers of TILs was studied in human melanoma tumors using patient-derived xenografts (n = 3 patients, n = 3 mice each), in AURKAi clinical trial samples (n = 3 patients, before/after therapy), and in The Cancer Genome Atlas (n = 278). All statistical tests were two-sided. RESULTS AURKAi response was associated with induction of the immune transcriptome (P = 3.5 x 10-29) while resistance inversely correlated with TIL numbers (Spearman r = -0.87, P < .001). AURKAi and CDK4/6i promoted the recruitment of TILs by inducing CCL5 secretion in melanoma cells (P ≤ .005) in an NF-κB-dependent manner. Therapeutic response to AURKAi was impaired in immunodeficient compared with immunocompetent mice (0% vs 67% tumors regressed, P = .01) and in mice bearing CCL5-deficient vs control tumors (P = .61 vs P = .02); however, AURKAi response was greatly enhanced in mice also receiving T-cell-activating immunotherapy (P < .001). In human tumors, CCL5 expression was also induced by AURKAi (P ≤ .02) and CDK4/6i (P = .01) and was associated with increased immune marker expression (P = 1.40 x 10-93). CONCLUSIONS Senescent melanoma cells secret CCL5, which promotes recruitment of TILs. Combining TIS with immunotherapy that enhances tumor cell killing by TILs is a promising novel approach to improve melanoma outcomes.


Journal of Investigative Dermatology | 2015

Acute Inhibition of MEK Suppresses Congenital Melanocytic Nevus Syndrome in a Murine Model Driven by Activated NRAS and Wnt Signaling

Jeff S. Pawlikowski; Claire Brock; Sheau-Chiann Chen; Lara Al-Olabi; Colin Nixon; Fiona McGregor; Simon Paine; Estelle Chanudet; Wendy Lambie; William M. Holmes; James Mullin; Ann Richmond; Hong Wu; Karen Blyth; Ayala King; V.A. Kinsler; Peter D. Adams

Congenital melanocytic nevus (CMN) syndrome is the association of pigmented melanocytic nevi with extra-cutaneous features, classically melanotic cells within the central nervous system, most frequently caused by a mutation of NRAS codon 61. This condition is currently untreatable and carries a significant risk of melanoma within the skin, brain, or leptomeninges. We have previously proposed a key role for Wnt signaling in the formation of melanocytic nevi, suggesting that activated Wnt signaling may be synergistic with activated NRAS in the pathogenesis of CMN syndrome. Some familial pre-disposition suggests a germ-line contribution to CMN syndrome, as does variability of neurological phenotypes in individuals with similar cutaneous phenotypes. Accordingly, we performed exome sequencing of germ-line DNA from patients with CMN to reveal rare or undescribed Wnt-signaling alterations. A murine model harboring activated NRASQ61K and Wnt signaling in melanocytes exhibited striking features of CMN syndrome, in particular neurological involvement. In the first model of treatment for this condition, these congenital, and previously assumed permanent, features were profoundly suppressed by acute post-natal treatment with a MEK inhibitor. These data suggest that activated NRAS and aberrant Wnt signaling conspire to drive CMN syndrome. Post-natal MEK inhibition is a potential candidate therapy for patients with this debilitating condition.


Clinical Cancer Research | 2015

Combining an Aurora Kinase Inhibitor and a Death Receptor Ligand/Agonist Antibody Triggers Apoptosis in Melanoma Cells and Prevents Tumor Growth in Preclinical Mouse Models

Yan Liu; Oriana E. Hawkins; Anna E. Vilgelm; Jeff S. Pawlikowski; Jeffrey Ecsedy; Jeffrey A. Sosman; Mark C. Kelley; Ann Richmond

Purpose: Preclinical studies show that inhibition of aurora kinases in melanoma tumors induces senescence and reduces tumor growth, but does not cause tumor regression. Additional preclinical models are needed to identify agents that will synergize with aurora kinase inhibitors to induce tumor regression. Experimental Design: We combined treatment with an aurora kinase A inhibitor, MLN8237, with agents that activate death receptors (Apo2L/TRAIL or death receptor 5 agonists) and monitored the ability of this treatment to induce tumor apoptosis and melanoma tumor regression using human cell lines and patient-derived xenograft (PDX) mouse models. Results: We found that this combined treatment led to apoptosis and markedly reduced cell viability. Mechanistic analysis showed that the induction of tumor cell senescence in response to the AURKA inhibitor resulted in a decreased display of Apo2L/TRAIL decoy receptors and increased display of one Apo2L/TRAIL receptor (death receptor 5), resulting in enhanced response to death receptor ligand/agonists. When death receptors were activated in senescent tumor cells, both intrinsic and extrinsic apoptotic pathways were induced independent of BRAF, NRAS, or p53 mutation status. Senescent tumor cells exhibited BID-mediated mitochondrial depolarization in response to Apo2L/TRAIL treatment. In addition, senescent tumor cells had a lower apoptotic threshold due to decreased XIAP and survivin expression. Melanoma tumor xenografts of one human cell line and one PDX displayed total blockage of tumor growth when treated with MLN8237 combined with DR5 agonist antibody. Conclusions: These findings provide a strong rationale for combining senescence-inducing therapeutics with death receptor agonists for improved cancer treatment. Clin Cancer Res; 21(23); 5338–48. ©2015 AACR.


Journal of Biosciences | 2007

A model for cell type localization in the migrating slug of Dictyostelium discoideum based on differential chemotactic sensitivity to cAMP and differential sensitivity to suppression of chemotaxis by ammonia

Ira N. Feit; Jeff S. Pawlikowski; Caroline Zawilski

The three basic cell types in the migrating slug of Dictyostelium discoideum show differential chemotactic response to cyclic AMP (cAMP) and differential sensitivity to suppression of the chemotaxis by ammonia. The values of these parameters indicate a progressive maturation of chemotactic properties during the transdifferentiation of slug cell types. We present a model that explains the localization of the three cell types within the slug based on these chemotactic differences and on the maturation of their chemotactic properties.

Collaboration


Dive into the Jeff S. Pawlikowski's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hong Wu

Fox Chase Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jeffrey Ecsedy

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge