Network


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

Hotspot


Dive into the research topics where Mark R. Woodford is active.

Publication


Featured researches published by Mark R. Woodford.


Molecular Cell | 2014

Asymmetric Hsp90 N Domain SUMOylation Recruits Aha1 and ATP-Competitive Inhibitors

Mehdi Mollapour; Dimitra Bourboulia; Kristin Beebe; Mark R. Woodford; Sigrun Polier; Anthony N. Hoang; Raju Chelluri; Yu Li; Ailan Guo; Min-Jung Lee; Elham Fotooh-Abadi; Sahar Khan; Thomas Prince; Naoto Miyajima; Soichiro Yoshida; Shinji Tsutsumi; Wanping Xu; Barry Panaretou; William G. Stetler-Stevenson; Gennady Bratslavsky; Jane B. Trepel; Chrisostomos Prodromou; Len Neckers

The stability and activity of numerous signaling proteins in both normal and cancer cells depends on the dimeric molecular chaperone heat shock protein 90 (Hsp90). Hsp90s function is coupled to ATP binding and hydrolysis and requires a series of conformational changes that are regulated by cochaperones and numerous posttranslational modifications (PTMs). SUMOylation is one of the least-understood Hsp90 PTMs. Here, we show that asymmetric SUMOylation of a conserved lysine residue in the N domain of both yeast (K178) and human (K191) Hsp90 facilitates both recruitment of the adenosine triphosphatase (ATPase)-activating cochaperone Aha1 and, unexpectedly, the binding of Hsp90 inhibitors, suggesting that these drugs associate preferentially with Hsp90 proteins that are actively engaged in the chaperone cycle. Importantly, cellular transformation is accompanied by elevated steady-state N domain SUMOylation, and increased Hsp90 SUMOylation sensitizes yeast and mammalian cells to Hsp90 inhibitors, providing a mechanism to explain the sensitivity of cancer cells to these drugs.


Cell Reports | 2015

c-Abl Mediated Tyrosine Phosphorylation of Aha1 Activates Its Co-chaperone Function in Cancer Cells

Diana M. Dunn; Mark R. Woodford; Andrew W. Truman; Sandra M. Jensen; Jacqualyn Schulman; Tiffany Caza; Taylor C. Remillard; David Loiselle; Don Wolfgeher; Brian S. J. Blagg; Lucas S. Franco; Timothy A. J. Haystead; Soumya Daturpalli; Matthias P. Mayer; Jane B. Trepel; Rhodri M. L. Morgan; Chrisostomos Prodromou; Stephen J. Kron; Barry Panaretou; William G. Stetler-Stevenson; Steve K. Landas; Len Neckers; Gennady Bratslavsky; Dimitra Bourboulia; Mehdi Mollapour

Summary The ability of Heat Shock Protein 90 (Hsp90) to hydrolyze ATP is essential for its chaperone function. The co-chaperone Aha1 stimulates Hsp90 ATPase activity, tailoring the chaperone function to specific “client” proteins. The intracellular signaling mechanisms directly regulating Aha1 association with Hsp90 remain unknown. Here, we show that c-Abl kinase phosphorylates Y223 in human Aha1 (hAha1), promoting its interaction with Hsp90. This, consequently, results in an increased Hsp90 ATPase activity, enhances Hsp90 interaction with kinase clients, and compromises the chaperoning of non-kinase clients such as glucocorticoid receptor and CFTR. Suggesting a regulatory paradigm, we also find that Y223 phosphorylation leads to ubiquitination and degradation of hAha1 in the proteasome. Finally, pharmacologic inhibition of c-Abl prevents hAha1 interaction with Hsp90, thereby hypersensitizing cancer cells to Hsp90 inhibitors both in vitro and ex vivo.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Structural and functional basis of protein phosphatase 5 substrate specificity

Jasmeen Oberoi; Diana M. Dunn; Mark R. Woodford; Laura Mariotti; Jacqualyn Schulman; Dimitra Bourboulia; Mehdi Mollapour; Cara K. Vaughan

Significance The activity of many proteins is dependent on molecular chaperones and their accessory proteins, cochaperones. The ability of a cohort of kinases, many of which are oncogenic, to transduce signals is promoted by the heat shock protein 90 (Hsp90) chaperone and Cdc37 cochaperone, and requires the removal of a phosphate from Cdc37 by protein phosphatase 5 (PP5). We present the crystal structure of PP5 with Cdc37 trapped in the active site. The structure reveals how PP5 can associate with different substrates and previously unknown determinants of specificity. Our findings show kinase release from the chaperone complex is critically dependent on this dephosphorylation and that combined inhibition of both Hsp90 and PP5 could provide an effective therapeutic strategy for treating cancers addicted to these kinases. The serine/threonine phosphatase protein phosphatase 5 (PP5) regulates hormone- and stress-induced cellular signaling by association with the molecular chaperone heat shock protein 90 (Hsp90). PP5-mediated dephosphorylation of the cochaperone Cdc37 is essential for activation of Hsp90-dependent kinases. However, the details of this mechanism remain unknown. We determined the crystal structure of a Cdc37 phosphomimetic peptide bound to the catalytic domain of PP5. The structure reveals PP5 utilization of conserved elements of phosphoprotein phosphatase (PPP) structure to bind substrate and provides a template for many PPP–substrate interactions. Our data show that, despite a highly conserved structure, elements of substrate specificity are determined within the phosphatase catalytic domain itself. Structure-based mutations in vivo reveal that PP5-mediated dephosphorylation is required for kinase and steroid hormone receptor release from the chaperone complex. Finally, our data show that hyper- or hypoactivity of PP5 mutants increases Hsp90 binding to its inhibitor, suggesting a mechanism to enhance the efficacy of Hsp90 inhibitors by regulation of PP5 activity in tumors.


Cell Reports | 2016

Mps1 Mediated Phosphorylation of Hsp90 Confers Renal Cell Carcinoma Sensitivity and Selectivity to Hsp90 Inhibitors

Mark R. Woodford; Andrew W. Truman; Diana M. Dunn; Sandra M. Jensen; Richard Cotran; Renee Bullard; Mourad Abouelleil; Kristin Beebe; Don Wolfgeher; Sara Wierzbicki; Dawn E. Post; Tiffany Caza; Shinji Tsutsumi; Barry Panaretou; Stephen J. Kron; Jane B. Trepel; Steve K. Landas; Chrisostomos Prodromou; Oleg Shapiro; William G. Stetler-Stevenson; Dimitra Bourboulia; Len Neckers; Gennady Bratslavsky; Mehdi Mollapour

SUMMARY The molecular chaperone Hsp90 protects deregulated signaling proteins that are vital for tumor growth and survival. Tumors generally display sensitivity and selectivity toward Hsp90 inhibitors; however, the molecular mechanism underlying this phenotype remains undefined. We report that the mitotic checkpoint kinase Mps1 phosphorylates a conserved threonine residue in the amino-domain of Hsp90. This, in turn, regulates chaperone function by reducing Hsp90 ATPase activity while fostering Hsp90 association with kinase clients, including Mps1. Phosphorylation of Hsp90 is also essential for the mitotic checkpoint because it confers Mps1 stability and activity. We identified Cdc14 as the phosphatase that dephosphorylates Hsp90 and disrupts its interaction with Mps1. This causes Mps1 degradation, thus providing a mechanism for its inactivation. Finally, Hsp90 phosphorylation sensitizes cells to its inhibitors, and elevated Mps1 levels confer renal cell carcinoma selectivity to Hsp90 drugs. Mps1 expression level can potentially serve as a predictive indicator of tumor response to Hsp90 inhibitors.


Nature Communications | 2016

The FNIP co-chaperones decelerate the Hsp90 chaperone cycle and enhance drug binding

Mark R. Woodford; Diana M. Dunn; Adam R. Blanden; Dante Capriotti; David Loiselle; Chrisostomos Prodromou; Barry Panaretou; Philip F. Hughes; Aaron P. Smith; Wendi Ackerman; Timothy A. J. Haystead; Stewart N. Loh; Dimitra Bourboulia; Laura S. Schmidt; W. Marston Linehan; Gennady Bratslavsky; Mehdi Mollapour

Heat shock protein-90 (Hsp90) is an essential molecular chaperone in eukaryotes involved in maintaining the stability and activity of numerous signalling proteins, also known as clients. Hsp90 ATPase activity is essential for its chaperone function and it is regulated by co-chaperones. Here we show that the tumour suppressor FLCN is an Hsp90 client protein and its binding partners FNIP1/FNIP2 function as co-chaperones. FNIPs decelerate the chaperone cycle, facilitating FLCN interaction with Hsp90, consequently ensuring FLCN stability. FNIPs compete with the activating co-chaperone Aha1 for binding to Hsp90, thereby providing a reciprocal regulatory mechanism for chaperoning of client proteins. Lastly, downregulation of FNIPs desensitizes cancer cells to Hsp90 inhibitors, whereas FNIPs overexpression in renal tumours compared with adjacent normal tissues correlates with enhanced binding of Hsp90 to its inhibitors. Our findings suggest that FNIPs expression can potentially serve as a predictive indicator of tumour response to Hsp90 inhibitors.


The EMBO Journal | 2017

Tumor suppressor Tsc1 is a new Hsp90 co‐chaperone that facilitates folding of kinase and non‐kinase clients

Mark R. Woodford; Rebecca A. Sager; Elijah Marris; Diana M. Dunn; Adam R. Blanden; Ryan L Murphy; Nicholas Rensing; Oleg Shapiro; Barry Panaretou; Chrisostomos Prodromou; Stewart N. Loh; David H. Gutmann; Dimitra Bourboulia; Gennady Bratslavsky; Michael Wong; Mehdi Mollapour

The tumor suppressors Tsc1 and Tsc2 form the tuberous sclerosis complex (TSC), a regulator of mTOR activity. Tsc1 stabilizes Tsc2; however, the precise mechanism involved remains elusive. The molecular chaperone heat‐shock protein 90 (Hsp90) is an essential component of the cellular homeostatic machinery in eukaryotes. Here, we show that Tsc1 is a new co‐chaperone for Hsp90 that inhibits its ATPase activity. The C‐terminal domain of Tsc1 (998–1,164 aa) forms a homodimer and binds to both protomers of the Hsp90 middle domain. This ensures inhibition of both subunits of the Hsp90 dimer and prevents the activating co‐chaperone Aha1 from binding the middle domain of Hsp90. Conversely, phosphorylation of Aha1‐Y223 increases its affinity for Hsp90 and displaces Tsc1, thereby providing a mechanism for equilibrium between binding of these two co‐chaperones to Hsp90. Our findings establish an active role for Tsc1 as a facilitator of Hsp90‐mediated folding of kinase and non‐kinase clients—including Tsc2—thereby preventing their ubiquitination and proteasomal degradation.


Current Topics in Medicinal Chemistry | 2016

Targeting Hsp90 in Non-Cancerous Maladies

Mark R. Woodford; Diana M. Dunn; Joseph G. Ciciarelli; Kristin Beebe; Len Neckers; Mehdi Mollapour

Heat shock protein-90 (Hsp90) is a molecular chaperone critical to the folding, stability and activity of over 200 client proteins including many responsible for tumor initiation, progression and metastasis. Hsp90 chaperone function is linked to its ATPase activity and Hsp90 inhibitors interfere with this activity, thereby making Hsp90 an attractive target for cancer therapy. Also post-translational modification (PTM) and co-chaperone proteins modulate Hsp90 function, providing additional targets for secondary inhibition. Recent reports have shown that pathogens utilize both their own Hsp90 and that of their host for the propagation of infectious elements. In this review we will summarize our current knowledge of Hsp90 structure and function in both the pathogen and the host. We will focus on the role of Hsp90 in viral and parasitic diseases and the potential beneficial application of Hsp90 inhibitors alone and in combination with disease-specific inhibitors.


Journal of Clinical Neuroscience | 2014

CIP2A and PP2A in human leptomeninges, arachnoid granulations and meningiomas

Mahlon D. Johnson; Jay E. Reeder; Mary O’Connell; Mark R. Woodford; Kevin A. Walter

Previously we have found that mitogens stimulate proliferation of meningioma cells of all grades, in part, by activation of the PI3K-PKB/Akt-PRAS40-mTOR pathway regulated to some degree by the tumor suppressor phosphatase PP2A. PP2A activity is inhibited by the oncoprotein cancerous inhibitor of PP2A (CIP2A), which has not been studied in meningiomas to our knowledge. Six fetal and one adult human leptomeningeal samples and 38 meningiomas were evaluated by western blot. Fifteen adult arachnoid granulations and 58 formalin-fixed meningiomas (36 World Health Organization grade I, 15 grade II and seven grade III) were evaluated by immunohistochemistry. The effects of the mitogens platelet derived growth factor-BB (PDGF-BB) and cerebrospinal fluid on CIP2A were also studied. By western blot, CIP2A and PP2A were found in the five fetal and one adult leptomeninges and all meningiomas. By immunohistochemistry, CIP2A was detected in the arachnoid granulations and all meningiomas. CIP2A tended to be higher in grade III tumors. Three fetal leptomeningeal (two grade I and one grade II) and meningioma cells treated with PDGF-BB and/or human cerebrospinal fluid resulted in a slight increase in CIP2A in the leptomeningeal cells but not meningioma cells. Considered the mechanism of action and seen in other neoplasms, these findings raise the possibility that CIP2A may participate in the biology of meningiomas.


Oncotarget | 2018

Sporadic renal angiomyolipoma in a patient with Birt-Hogg-Dubé: chaperones in pathogenesis

Rebecca A. Sager; Mark R. Woodford; Oleg Shapiro; Mehdi Mollapour; Gennady Bratslavsky

Birt-Hogg-Dubé (BHD) is an autosomal dominant genetic syndrome caused by germline mutations in the FLCN gene that predisposes patients to develop renal tumors. Renal angiomyolipoma (AML) is not a renal tumor sub-type associated with BHD. AML is, however, a common phenotypic manifestation of Tuberous Sclerosis Complex (TSC) syndrome caused by mutations in either the TSC1 or TSC2 tumor suppressor genes. Previous case reports of renal AML in patients with BHD have speculated on the molecular and clinical overlap of these two syndromes as a result of described involvement of the gene products in the mTOR pathway. Our recent work provided a new molecular link between these two syndromes by identifying FLCN and Tsc2 as clients of the molecular chaperone Hsp90. Folliculin interacting proteins FNIP1/2 and Tsc1 are important for FLCN and Tsc2 stability as new Hsp90 co-chaperones. Here we present a case of sporadic AML as a result of somatic Tsc1/2 loss in a patient with BHD. We further demonstrate that FNIP1 and Tsc1 are capable of compensating for each other in the chaperoning of mutated FLCN tumor suppressor. Our findings demonstrate interconnectivity and compensatory mechanisms between the BHD and TSC pathways.


Oncotarget | 2016

Sixth BHD Symposium and First International Upstate Kidney Cancer Symposium: latest scientific and clinical discoveries

Gennady Bratslavsky; Mark R. Woodford; Michael Daneshvar; Mehdi Mollapour

The Sixth BHD Symposium and First International Upstate Kidney Cancer Symposium concluded in September 2015, in Syracuse, NY, USA. The program highlighted recent findings in a variety of areas, including drug development, therapeutics and surgical management of patients with BHD and multi-focal renal tumors, as well as multidisciplinary approaches for patients with localized, locally advanced and metastatic renal cell carcinoma.

Collaboration


Dive into the Mark R. Woodford's collaboration.

Top Co-Authors

Avatar

Mehdi Mollapour

State University of New York Upstate Medical University

View shared research outputs
Top Co-Authors

Avatar

Gennady Bratslavsky

State University of New York Upstate Medical University

View shared research outputs
Top Co-Authors

Avatar

Dimitra Bourboulia

State University of New York Upstate Medical University

View shared research outputs
Top Co-Authors

Avatar

Diana M. Dunn

State University of New York Upstate Medical University

View shared research outputs
Top Co-Authors

Avatar

Len Neckers

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Rebecca A. Sager

State University of New York Upstate Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Oleg Shapiro

State University of New York Upstate Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge