Network


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

Hotspot


Dive into the research topics where Peter S. Winter is active.

Publication


Featured researches published by Peter S. Winter.


Journal of Clinical Investigation | 2015

Metabolic programming and PDHK1 control CD4+ T cell subsets and inflammation

Valerie A. Gerriets; Rigel J. Kishton; Amanda G. Nichols; Andrew N. Macintyre; Makoto Inoue; Olga Ilkayeva; Peter S. Winter; Xiaojing Liu; Bhavana Priyadharshini; Marta E. Slawinska; Lea Haeberli; Catherine Huck; Laurence A. Turka; Kris C. Wood; Laura P. Hale; Paul Smith; Martin A. Schneider; Nancie J. MacIver; Jason W. Locasale; Christopher B. Newgard; Mari L. Shinohara; Jeffrey C. Rathmell

Activation of CD4+ T cells results in rapid proliferation and differentiation into effector and regulatory subsets. CD4+ effector T cell (Teff) (Th1 and Th17) and Treg subsets are metabolically distinct, yet the specific metabolic differences that modify T cell populations are uncertain. Here, we evaluated CD4+ T cell populations in murine models and determined that inflammatory Teffs maintain high expression of glycolytic genes and rely on high glycolytic rates, while Tregs are oxidative and require mitochondrial electron transport to proliferate, differentiate, and survive. Metabolic profiling revealed that pyruvate dehydrogenase (PDH) is a key bifurcation point between T cell glycolytic and oxidative metabolism. PDH function is inhibited by PDH kinases (PDHKs). PDHK1 was expressed in Th17 cells, but not Th1 cells, and at low levels in Tregs, and inhibition or knockdown of PDHK1 selectively suppressed Th17 cells and increased Tregs. This alteration in the CD4+ T cell populations was mediated in part through ROS, as N-acetyl cysteine (NAC) treatment restored Th17 cell generation. Moreover, inhibition of PDHK1 modulated immunity and protected animals against experimental autoimmune encephalomyelitis, decreasing Th17 cells and increasing Tregs. Together, these data show that CD4+ subsets utilize and require distinct metabolic programs that can be targeted to control specific T cell populations in autoimmune and inflammatory diseases.


Science Signaling | 2014

Systematic identification of signaling pathways with potential to confer anticancer drug resistance.

Colin A. Martz; Kathleen Ottina; Katherine R. Singleton; Jeff S. Jasper; Suzanne E. Wardell; Ashley Peraza-Penton; Grace R. Anderson; Peter S. Winter; Tim Wang; Holly M. Alley; Lawrence N. Kwong; Zachary A. Cooper; Michael T. Tetzlaff; Pei Ling Chen; Jeffrey C. Rathmell; Keith T. Flaherty; Jennifer A. Wargo; Donald P. McDonnell; David M. Sabatini; Kris C. Wood

Pathway-centric screening reveals new mechanisms of drug resistance and combination therapeutic strategies. Finding New Targets for Drug-Resistant Cancers The development of drug resistance is a common problem in cancer patients. Knowing how drug resistance emerged in a tumor can inform clinical strategy. Martz et al. devised a drug screen to identify pathways of resistance when cancer cells were treated with drugs that are used in the clinic. Along with pathways known to mediate drug resistance, such as the MAPK and PI3K pathways, activation of the Notch1 pathway caused drug resistance in various types of cancer cells in culture. Inhibiting Notch1 signaling restored drug efficacy in cells in culture and in xenografts in mice. Intriguingly, Notch signaling mediated drug resistance to an estrogen receptor–targeted therapy used in breast cancer and to a kinase-targeted therapy used in melanoma, suggesting that this single pathway may be important in multiple types of drug-resistant cancers. Indeed, tumors of some patients with relapsed breast cancer or melanoma had increased markers of Notch1 signaling. In the Research Article by Winter et al. also in this issue, this screening method identified a pathway of drug resistance in bone marrow cancer. More generally, by screening entire signaling pathways instead of individual genes, the work of Martz et al. shows how we can quickly map pathways to the diverse properties of cancer cells. Cancer cells can activate diverse signaling pathways to evade the cytotoxic action of drugs. We created and screened a library of barcoded pathway-activating mutant complementary DNAs to identify those that enhanced the survival of cancer cells in the presence of 13 clinically relevant, targeted therapies. We found that activation of the RAS-MAPK (mitogen-activated protein kinase), Notch1, PI3K (phosphoinositide 3-kinase)–mTOR (mechanistic target of rapamycin), and ER (estrogen receptor) signaling pathways often conferred resistance to this selection of drugs. Activation of the Notch1 pathway promoted acquired resistance to tamoxifen (an ER-targeted therapy) in serially passaged breast cancer xenografts in mice, and treating mice with a γ-secretase inhibitor to inhibit Notch signaling restored tamoxifen sensitivity. Markers of Notch1 activity in tumor tissue correlated with resistance to tamoxifen in breast cancer patients. Similarly, activation of Notch1 signaling promoted acquired resistance to MAPK inhibitors in BRAFV600E melanoma cells in culture, and the abundance of Notch1 pathway markers was increased in tumors from a subset of melanoma patients. Thus, Notch1 signaling may be a therapeutic target in some drug-resistant breast cancers and melanomas. Additionally, multiple resistance pathways were activated in melanoma cell lines with intrinsic resistance to MAPK inhibitors, and simultaneous inhibition of these pathways synergistically induced drug sensitivity. These data illustrate the potential for systematic identification of the signaling pathways controlling drug resistance that could inform clinical strategies and drug development for multiple types of cancer. This approach may also be used to advance clinical options in other disease contexts.


Science Signaling | 2014

RAS signaling promotes resistance to JAK inhibitors by suppressing BAD-mediated apoptosis

Peter S. Winter; Kristopher A. Sarosiek; Kevin Lin; Manja Meggendorfer; Susanne Schnittger; Anthony Letai; Kris C. Wood

Targeting a protein that prevents cell death may overcome drug resistance in some blood cancers. Overcoming Resistance in Bone Marrow Cancer An activating mutation in the kinase JAK2 (Janus kinase 2) is common in patients with myeloproliferative neoplasms (MPNs), which are abnormal growths of cells in the bone marrow that may progress to acute myeloid leukemia. However, cells often show inherent resistance to clinically used JAK inhibitors. Using a pathway-centric screen described by Martz et al. in this issue, Winter et al. found that the pathway mediated by RAS, another protein that is frequently activated in MPNs and numerous other cancers, promoted resistance in hematopoietic cell lines containing an activating mutation in JAK2. RAS signaling led to the phosphorylation-mediated inactivation of the death-promoting (proapoptotic) protein BAD, enabling cell survival. Combining inhibitors of kinases downstream of RAS signaling with JAK inhibitors resulted in cell death of cultured cancer cell lines resistant to JAK inhibitors, as did an inhibitor of an antiapoptotic protein. The findings identify a potential therapeutic option for drug-resistant MPNs. Myeloproliferative neoplasms (MPNs) frequently have an activating mutation in the gene encoding Janus kinase 2 (JAK2). Thus, targeting the pathway mediated by JAK and its downstream substrate, signal transducer and activator of transcription (STAT), may yield clinical benefit for patients with MPNs containing the JAK2V617F mutation. Although JAK inhibitor therapy reduces splenomegaly and improves systemic symptoms in patients, this treatment does not appreciably reduce the number of neoplastic cells. To identify potential mechanisms underlying this inherent resistance phenomenon, we performed pathway-centric, gain-of-function screens in JAK2V617F hematopoietic cells and found that the activation of the guanosine triphosphatase (GTPase) RAS or its effector pathways [mediated by the kinases AKT and ERK (extracellular signal–regulated kinase)] renders cells insensitive to JAK inhibition. Resistant MPN cells became sensitized to JAK inhibitors when also exposed to inhibitors of the AKT or ERK pathways. Mechanistically, in JAK2V617F cells, a JAK2-mediated inactivating phosphorylation of the proapoptotic protein BAD [B cell lymphoma 2 (BCL-2)–associated death promoter] promoted cell survival. In sensitive cells, exposure to a JAK inhibitor resulted in dephosphorylation of BAD, enabling BAD to bind and sequester the prosurvival protein BCL-XL (BCL-2–like 1), thereby triggering apoptosis. In resistant cells, RAS effector pathways maintained BAD phosphorylation in the presence of JAK inhibitors, yielding a specific dependence on BCL-XL for survival. In patients with MPNs, activating mutations in RAS co-occur with the JAK2V617F mutation in the malignant cells, suggesting that RAS effector pathways likely play an important role in clinically observed resistance.


Scientific Reports | 2016

Targeting MCL-1/BCL-XL Forestalls the Acquisition of Resistance to ABT-199 in Acute Myeloid Leukemia

Kevin Lin; Peter S. Winter; Abigail Xie; Cullen Roth; Colin A. Martz; Elizabeth M. Stein; Grace R. Anderson; Jennifer P. Tingley; Kris C. Wood

ABT-199, a potent and selective small-molecule antagonist of BCL-2, is being clinically vetted as pharmacotherapy for the treatment of acute myeloid leukemia (AML). However, given that prolonged monotherapy tends to beget resistance, we sought to investigate the means by which resistance to ABT-199 might arise in AML and the extent to which those mechanisms might be preempted. Here we used a pathway-activating genetic screen to nominate MCL-1 and BCL-XL as potential nodes of resistance. We then characterized a panel of ABT-199-resistant myeloid leukemia cell lines derived through chronic exposure to ABT-199 and found that acquired drug resistance is indeed driven by the upregulation of MCL-1 and BCL-XL. By targeting MCL-1 and BCL-XL, resistant AML cell lines could be resensitized to ABT-199. Further, preemptively targeting MCL-1 and/or BCL-XL alongside administration of ABT-199 was capable of delaying or forestalling the acquisition of drug resistance. Collectively, these data suggest that in AML, (1) the selection of initial therapy dynamically templates the landscape of acquired resistance via modulation of MCL-1/BCL-XL and (2) appropriate selection of initial therapy may delay or altogether forestall the acquisition of resistance to ABT-199.


Genome Research | 2016

Mapping nucleosome positions using DNase-seq

Jianling Zhong; Kaixuan Luo; Peter S. Winter; Gregory E. Crawford; Edwin S. Iversen; Alexander J. Hartemink

Although deoxyribonuclease I (DNase I) was used to probe the structure of the nucleosome in the 1960s and 1970s, in the current high-throughput sequencing era, DNase I has mainly been used to study genomic regions devoid of nucleosomes. Here, we reveal for the first time that DNase I can be used to precisely map the (translational) positions of in vivo nucleosomes genome-wide. Specifically, exploiting a distinctive DNase I cleavage profile within nucleosome-associated DNA--including a signature 10.3 base pair oscillation that corresponds to accessibility of the minor groove as DNA winds around the nucleosome--we develop a Bayes-factor-based method that can be used to map nucleosome positions along the genome. Compared to methods that require genetically modified histones, our DNase-based approach is easily applied in any organism, which we demonstrate by producing maps in yeast and human. Compared to micrococcal nuclease (MNase)-based methods that map nucleosomes based on cuts in linker regions, we utilize DNase I cuts both outside and within nucleosomal DNA; the oscillatory nature of the DNase I cleavage profile within nucleosomal DNA enables us to identify translational positioning details not apparent in MNase digestion of linker DNA. Because the oscillatory pattern corresponds to nucleosome rotational positioning, it also reveals the rotational context of transcription factor (TF) binding sites. We show that potential binding sites within nucleosome-associated DNA are often centered preferentially on an exposed major or minor groove. This preferential localization may modulate TF interaction with nucleosome-associated DNA as TFs search for binding sites.


Cell Reports | 2017

A Landscape of Therapeutic Cooperativity in KRAS Mutant Cancers Reveals Principles for Controlling Tumor Evolution

Grace R. Anderson; Peter S. Winter; Kevin Lin; Daniel P. Nussbaum; Merve Cakir; Elizabeth M. Stein; Ryan S. Soderquist; Lorin Crawford; Jim C. Leeds; Rachel Newcomb; Priya Stepp; Catherine Yip; Suzanne E. Wardell; Jennifer P. Tingley; Moiez Ali; MengMeng Xu; Meagan Ryan; Shannon McCall; Autumn J. McRee; Christopher M. Counter; Channing J. Der; Kris C. Wood

Combinatorial inhibition of effector and feedback pathways is a promising treatment strategy for KRAS mutant cancers. However, the particular pathways that should be targeted to optimize therapeutic responses are unclear. Using CRISPR/Cas9, we systematically mapped the pathways whose inhibition cooperates with drugs targeting the KRAS effectors MEK, ERK, and PI3K. By performing 70 screens in models of KRAS mutant colorectal, lung, ovarian, and pancreas cancers, we uncovered universal and tissue-specific sensitizing combinations involving inhibitors of cell cycle, metabolism, growth signaling, chromatin regulation, and transcription. Furthermore, these screens revealed secondary genetic modifiers of sensitivity, yielding a SRC inhibitor-based combination therapy for KRAS/PIK3CA double-mutant colorectal cancers (CRCs) with clinical potential. Surprisingly, acquired resistance to combinations of growth signaling pathway inhibitors develops rapidly following treatment, but by targeting signaling feedback or apoptotic priming, it is possible to construct three-drug combinations that greatly delay its emergence.


PLOS ONE | 2014

Systemic acquired resistance in moss: further evidence for conserved defense mechanisms in plants.

Peter S. Winter; Collin E. Bowman; Philip Villani; Thomas E. Dolan; Nathanael R. Hauck

Vascular plants possess multiple mechanisms for defending themselves against pathogens. One well-characterized defense mechanism is systemic acquired resistance (SAR). In SAR, a plant detects the presence of a pathogen and transmits a signal throughout the plant, inducing changes in the expression of various pathogenesis-related (PR) genes. Once SAR is established, the plant is capable of mounting rapid responses to subsequent pathogen attacks. SAR has been characterized in numerous angiosperm and gymnosperm species; however, despite several pieces of evidence suggesting SAR may also exist in non-vascular plants6–8, its presence in non-vascular plants has not been conclusively demonstrated, in part due to the lack of an appropriate culture system. Here, we describe and use a novel culture system to demonstrate that the moss species Amblystegium serpens does initiate a SAR-like reaction upon inoculation with Pythium irregulare, a common soil-borne oomycete. Infection of A. serpens gametophores by P. irregulare is characterized by localized cytoplasmic shrinkage within 34 h and chlorosis and necrosis within 7 d of inoculation. Within 24 h of a primary inoculation (induction), moss gametophores grown in culture became highly resistant to infection following subsequent inoculation (challenge) by the same pathogen. This increased resistance was a response to the pathogen itself and not to physical wounding. Treatment with β-1,3 glucan, a structural component of oomycete cell walls, was equally effective at triggering SAR. Our results demonstrate, for the first time, that this important defense mechanism exists in a non-vascular plant, and, together with previous studies, suggest that SAR arose prior to the divergence of vascular and non-vascular plants. In addition, this novel moss – pathogen culture system will be valuable for future characterization of the mechanism of SAR in moss, which is necessary for a better understanding of the evolutionary history of SAR in plants.


Science Translational Medicine | 2016

PIK3CA mutations enable targeting of a breast tumor dependency through mTOR-mediated MCL-1 translation

Grace R. Anderson; Suzanne E. Wardell; Merve Cakir; Lorin Crawford; J Leeds; Daniel P. Nussbaum; Ps Shankar; Ryan S. Soderquist; Elizabeth M. Stein; Jennifer P. Tingley; Peter S. Winter; Ek Zieser-Misenheimer; Holly M. Alley; Alexander P. Yllanes; Haney; Kimberly L. Blackwell; Shannon McCall; Donald P. McDonnell; Kris C. Wood

Inhibitors of BCL-XL, combined with inhibition of the mTOR/4E-BP axis, drive regressions of PIK3CA mutant breast tumors. Sneak attack on breast cancer’s defense The usual goal of cancer treatment is to kill malignant cells, not just slow down their growth. A class of drugs called BH3 mimetics serves this purpose by inhibiting antiapoptotic proteins and thus helping drive the cells toward apoptosis (programmed cell death). MCL-1 is an antiapoptotic protein that is not targeted by currently bioavailable BH3 mimetics, and it is often responsible for resistance to these drugs. Anderson et al. have discovered that breast cancers with the commonly observed PIK3CA mutations can be treated with mTOR inhibitors to suppress MCL-1, leaving the cells vulnerable to BH3 mimetics and subsequent induction of apoptosis, both directly and in combination with chemotherapy. Therapies that efficiently induce apoptosis are likely to be required for durable clinical responses in patients with solid tumors. Using a pharmacological screening approach, we discovered that combined inhibition of B cell lymphoma–extra large (BCL-XL) and the mammalian target of rapamycin (mTOR)/4E-BP axis results in selective and synergistic induction of apoptosis in cellular and animal models of PIK3CA mutant breast cancers, including triple-negative tumors. Mechanistically, inhibition of mTOR/4E-BP suppresses myeloid cell leukemia–1 (MCL-1) protein translation only in PIK3CA mutant tumors, creating a synthetic dependence on BCL-XL. This dual dependence on BCL-XL and MCL-1, but not on BCL-2, appears to be a fundamental property of diverse breast cancer cell lines, xenografts, and patient-derived tumors that is independent of the molecular subtype or PIK3CA mutational status. Furthermore, this dependence distinguishes breast cancers from normal breast epithelial cells, which are neither primed for apoptosis nor dependent on BCL-XL/MCL-1, suggesting a potential therapeutic window. By tilting the balance of pro- to antiapoptotic signals in the mitochondria, dual inhibition of MCL-1 and BCL-XL also sensitizes breast cancer cells to standard-of-care cytotoxic and targeted chemotherapies. Together, these results suggest that patients with PIK3CA mutant breast cancers may benefit from combined treatment with inhibitors of BCL-XL and the mTOR/4E-BP axis, whereas alternative methods of inhibiting MCL-1 and BCL-XL may be effective in tumors lacking PIK3CA mutations.


eLife | 2017

Epstein-Barr virus ensures B cell survival by uniquely modulating apoptosis at early and late times after infection

Alexander M. Price; Joanne Dai; Quentin Bazot; Luv Patel; Pavel A. Nikitin; Reza Djavadian; Peter S. Winter; Cristina A Salinas; Ashley Perkins Barry; Kris C. Wood; Eric Johannsen; Anthony Letai; Martin J. Allday; Micah A. Luftig

Latent Epstein-Barr virus (EBV) infection is causally linked to several human cancers. EBV expresses viral oncogenes that promote cell growth and inhibit the apoptotic response to uncontrolled proliferation. The EBV oncoprotein LMP1 constitutively activates NFκB and is critical for survival of EBV-immortalized B cells. However, during early infection EBV induces rapid B cell proliferation with low levels of LMP1 and little apoptosis. Therefore, we sought to define the mechanism of survival in the absence of LMP1/NFκB early after infection. We used BH3 profiling to query mitochondrial regulation of apoptosis and defined a transition from uninfected B cells (BCL-2) to early-infected (MCL-1/BCL-2) and immortalized cells (BFL-1). This dynamic change in B cell survival mechanisms is unique to virus-infected cells and relies on regulation of MCL-1 mitochondrial localization and BFL-1 transcription by the viral EBNA3A protein. This study defines a new role for EBNA3A in the suppression of apoptosis with implications for EBV lymphomagenesis. DOI: http://dx.doi.org/10.7554/eLife.22509.001


Molecular Cancer Research | 2017

CDK4/6 Therapeutic Intervention and Viable Alternative to Taxanes in CRPC

James P. Stice; Suzanne E. Wardell; John D. Norris; Alexander P. Yllanes; Holly M. Alley; Victoria O. Haney; Hannah S. White; Rachid Safi; Peter S. Winter; Kimberly J. Cocce; Rigel J. Kishton; Scott A. Lawrence; Jay C. Strum; Donald P. McDonnell

Resistance to second-generation androgen receptor (AR) antagonists and CYP17 inhibitors in patients with castration-resistant prostate cancer (CRPC) develops rapidly through reactivation of the androgen signaling axis and has been attributed to AR overexpression, production of constitutively active AR splice variants, or the selection for AR mutants with altered ligand-binding specificity. It has been established that androgens induce cell-cycle progression, in part, through upregulation of cyclin D1 (CCND1) expression and subsequent activation of cyclin-dependent kinases 4 and 6 (CDK4/6). Thus, the efficacy of the newly described CDK4/6 inhibitors (G1T28 and G1T38), docetaxel and enzalutamide, was evaluated as single agents in clinically relevant in vitro and in vivo models of hormone-sensitive and treatment-resistant prostate cancer. CDK4/6 inhibition (CDK4/6i) was as effective as docetaxel in animal models of treatment-resistant CRPC but exhibited significantly less toxicity. The in vivo effects were durable and importantly were observed in prostate cancer cells expressing wild-type AR, AR mutants, and those that have lost AR expression. CDK4/6i was also effective in prostate tumor models expressing the AR-V7 variant or the AR F876L mutation, both of which are associated with treatment resistance. Furthermore, CDK4/6i was effective in prostate cancer models where AR expression was lost. It is concluded that CDK4/6 inhibitors are a viable alternative to taxanes as therapeutic interventions in endocrine therapy–refractory CRPC. Implications: The preclinical efficacy of CDK4/6 monotherapy observed here suggests the need for near-term clinical studies of these agents in advanced prostate cancer. Mol Cancer Res; 15(6); 660–9. ©2017 AACR.

Collaboration


Dive into the Peter S. Winter'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

Jeffrey C. Rathmell

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Kevin Lin

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge