Try a new search

Format these results:

Searched for:

person:kanshe01

in-biosketch:yes

Total Results:

29


Pilot study evaluating everolimus molecular mechanisms in tuberous sclerosis complex and focal cortical dysplasia

Leitner, Dominique F; Kanshin, Evgeny; Askenazi, Manor; Siu, Yik; Friedman, Daniel; Devore, Sasha; Jones, Drew; Ueberheide, Beatrix; Wisniewski, Thomas; Devinsky, Orrin
BACKGROUND:Tuberous sclerosis complex (TSC) and some focal cortical dysplasias (FCDs) are associated with dysfunctional mTOR signaling, resulting in increased cell growth and ribosomal S6 protein phosphorylation (phospho-S6). mTOR inhibitors can reduce TSC tumor growth and seizure frequency, and preclinical FCD studies indicate seizure suppression. This pilot study evaluated safety of mTOR inhibitor everolimus in treatment resistant (failure of >2 anti-seizure medications) TSC and FCD patients undergoing surgical resection and to assess mTOR signaling and molecular pathways. METHODS AND FINDINGS/RESULTS:We evaluated everolimus in 14 treatment resistant epilepsy patients undergoing surgical resection (4.5 mg/m2 daily for 7 days; n = 4 Active, mean age 18.3 years, range 4-26; n = 10, Control, mean age 13.1, range 3-45). Everolimus was well tolerated. Mean plasma everolimus in Active participants were in target range (12.4 ng/ml). Brain phospho-S6 was similar in Active and Control participants with a lower trend in Active participants, with Ser235/236 1.19-fold (p = 0.67) and Ser240/244 1.15-fold lower (p = 0.66). Histologically, Ser235/236 was 1.56-fold (p = 0.37) and Ser240/244 was 5.55-fold lower (p = 0.22). Brain proteomics identified 11 proteins at <15% false discovery rate associated with coagulation system (p = 1.45x10-9) and acute phase response (p = 1.23x10-6) activation. A weighted gene correlation network analysis (WGCNA) of brain proteomics and phospho-S6 identified 5 significant modules. Higher phospho-S6 correlated negatively with cellular respiration and synaptic transmission and positively with organophosphate metabolic process, nuclear mRNA catabolic process, and neuron ensheathment. Brain metabolomics identified 14 increased features in Active participants, including N-acetylaspartylglutamic acid. Plasma proteomics and cytokine analyses revealed no differences. CONCLUSIONS:Short-term everolimus before epilepsy surgery in TSC and FCD resulted in no adverse events and trending lower mTOR signaling (phospho-S6). Future studies should evaluate implications of our findings, including coagulation system activation and everolimus efficacy in FCD, in larger studies with long-term treatment to better understand molecular and clinical effects. CLINICAL TRIALS REGISTRATION/BACKGROUND:ClinicalTrials.gov NCT02451696.
PMCID:9119437
PMID: 35587487
ISSN: 1932-6203
CID: 5228952

Raphe and ventrolateral medulla proteomics in epilepsy and sudden unexpected death in epilepsy

Leitner, Dominique F; Kanshin, Evgeny; Askenazi, Manor; Faustin, Arline; Friedman, Daniel; Devore, Sasha; Ueberheide, Beatrix; Wisniewski, Thomas; Devinsky, Orrin
Brainstem nuclei dysfunction is implicated in sudden unexpected death in epilepsy. In animal models, deficient serotonergic activity is associated with seizure-induced respiratory arrest. In humans, glia are decreased in the ventrolateral medullary pre-Botzinger complex that modulate respiratory rhythm, as well as in the medial medullary raphe that modulate respiration and arousal. Finally, sudden unexpected death in epilepsy cases have decreased midbrain volume. To understand the potential role of brainstem nuclei in sudden unexpected death in epilepsy, we evaluated molecular signalling pathways using localized proteomics in microdissected midbrain dorsal raphe and medial medullary raphe serotonergic nuclei, as well as the ventrolateral medulla in brain tissue from epilepsy patients who died of sudden unexpected death in epilepsy and other causes in diverse epilepsy syndromes and non-epilepsy control cases (n = 15-16 cases per group/region). Compared with the dorsal raphe of non-epilepsy controls, we identified 89 proteins in non-sudden unexpected death in epilepsy and 219 proteins in sudden unexpected death in epilepsy that were differentially expressed. These proteins were associated with inhibition of EIF2 signalling (P-value of overlap = 1.29 × 10-8, z = -2.00) in non-sudden unexpected death in epilepsy. In sudden unexpected death in epilepsy, there were 10 activated pathways (top pathway: gluconeogenesis I, P-value of overlap = 3.02 × 10-6, z = 2.24) and 1 inhibited pathway (fatty acid beta-oxidation, P-value of overlap = 2.69 × 10-4, z = -2.00). Comparing sudden unexpected death in epilepsy and non-sudden unexpected death in epilepsy, 10 proteins were differentially expressed, but there were no associated signalling pathways. In both medullary regions, few proteins showed significant differences in pairwise comparisons. We identified altered proteins in the raphe and ventrolateral medulla of epilepsy patients, including some differentially expressed in sudden unexpected death in epilepsy cases. Altered signalling pathways in the dorsal raphe of sudden unexpected death in epilepsy indicate a shift in cellular energy production and activation of G-protein signalling, inflammatory response, stress response and neuronal migration/outgrowth. Future studies should assess the brain proteome in relation to additional clinical variables (e.g. recent tonic-clonic seizures) and in more of the reciprocally connected cortical and subcortical regions to better understand the pathophysiology of epilepsy and sudden unexpected death in epilepsy.
PMCID:9344977
PMID: 35928051
ISSN: 2632-1297
CID: 5288272

Proteomics and Transcriptomics of the Hippocampus and Cortex in SUDEP and High-Risk SUDEP Patients

Leitner, Dominique F; Mills, James D; Pires, Geoffrey; Faustin, Arline; Drummond, Eleanor; Kanshin, Evgeny; Nayak, Shruti; Askenazi, Manor; Verducci, Chloe; Chen, Bei Jun; Janitz, Michael; Anink, Jasper J; Baayen, Johannes C; Idema, Sander; van Vliet, Erwin A; Devore, Sasha; Friedman, Daniel; Diehl, Beate; Scott, Catherine; Thijs, Roland; Wisniewski, Thomas; Ueberheide, Beatrix; Thom, Maria; Aronica, Eleonora; Devinsky, Orrin
OBJECTIVE:To identify the molecular signaling pathways underlying sudden unexpected death in epilepsy (SUDEP) and high-risk SUDEP compared to epilepsy control patients. METHODS:For proteomics analyses, we evaluated the hippocampus and frontal cortex from microdissected post-mortem brain tissue of 12 SUDEP and 14 non-SUDEP epilepsy patients. For transcriptomics analyses, we evaluated hippocampus and temporal cortex surgical brain tissue from mesial temporal lobe epilepsy (MTLE) patients: 6 low-risk and 8 high-risk SUDEP as determined by a short (< 50 seconds) or prolonged (≥ 50 seconds) postictal generalized EEG suppression (PGES) that may indicate severely depressed brain activity impairing respiration, arousal, and protective reflexes. RESULTS:In autopsy hippocampus and cortex, we observed no proteomic differences between SUDEP and non-SUDEP epilepsy patients, contrasting with our previously reported robust differences between epilepsy and non-epilepsy control patients. Transcriptomics in hippocampus and cortex from surgical epilepsy patients segregated by PGES identified 55 differentially expressed genes (37 protein-coding, 15 lncRNAs, three pending) in hippocampus. CONCLUSION/CONCLUSIONS:The SUDEP proteome and high-risk SUDEP transcriptome were similar to other epilepsy patients in hippocampus and frontal cortex, consistent with diverse epilepsy syndromes and comorbidities associated with SUDEP. Studies with larger cohorts and different epilepsy syndromes, as well as additional anatomic regions may identify molecular mechanisms of SUDEP.
PMID: 33910938
ISSN: 1526-632x
CID: 4852152

PRMT5 inhibition disrupts splicing and stemness in glioblastoma

Sachamitr, Patty; Ho, Jolene C; Ciamponi, Felipe E; Ba-Alawi, Wail; Coutinho, Fiona J; Guilhamon, Paul; Kushida, Michelle M; Cavalli, Florence M G; Lee, Lilian; Rastegar, Naghmeh; Vu, Victoria; Sánchez-Osuna, María; Coulombe-Huntington, Jasmin; Kanshin, Evgeny; Whetstone, Heather; Durand, Mathieu; Thibault, Philippe; Hart, Kirsten; Mangos, Maria; Veyhl, Joseph; Chen, Wenjun; Tran, Nhat; Duong, Bang-Chi; Aman, Ahmed M; Che, Xinghui; Lan, Xiaoyang; Whitley, Owen; Zaslaver, Olga; Barsyte-Lovejoy, Dalia; Richards, Laura M; Restall, Ian; Caudy, Amy; Röst, Hannes L; Bonday, Zahid Quyoom; Bernstein, Mark; Das, Sunit; Cusimano, Michael D; Spears, Julian; Bader, Gary D; Pugh, Trevor J; Tyers, Mike; Lupien, Mathieu; Haibe-Kains, Benjamin; Artee Luchman, H; Weiss, Samuel; Massirer, Katlin B; Prinos, Panagiotis; Arrowsmith, Cheryl H; Dirks, Peter B
Glioblastoma (GBM) is a deadly cancer in which cancer stem cells (CSCs) sustain tumor growth and contribute to therapeutic resistance. Protein arginine methyltransferase 5 (PRMT5) has recently emerged as a promising target in GBM. Using two orthogonal-acting inhibitors of PRMT5 (GSK591 or LLY-283), we show that pharmacological inhibition of PRMT5 suppresses the growth of a cohort of 46 patient-derived GBM stem cell cultures, with the proneural subtype showing greater sensitivity. We show that PRMT5 inhibition causes widespread disruption of splicing across the transcriptome, particularly affecting cell cycle gene products. We identify a GBM splicing signature that correlates with the degree of response to PRMT5 inhibition. Importantly, we demonstrate that LLY-283 is brain-penetrant and significantly prolongs the survival of mice with orthotopic patient-derived xenografts. Collectively, our findings provide a rationale for the clinical development of brain penetrant PRMT5 inhibitors as treatment for GBM.
PMCID:7881162
PMID: 33579912
ISSN: 2041-1723
CID: 5228942

Proteomic differences in the hippocampus and cortex of epilepsy brain tissue

Pires, Geoffrey; Leitner, Dominique; Drummond, Eleanor; Kanshin, Evgeny; Nayak, Shruti; Askenazi, Manor; Faustin, Arline; Friedman, Daniel; Debure, Ludovic; Ueberheide, Beatrix; Wisniewski, Thomas; Devinsky, Orrin
Epilepsy is a common neurological disorder affecting over 70 million people worldwide, with a high rate of pharmaco-resistance, diverse comorbidities including progressive cognitive and behavioural disorders, and increased mortality from direct (e.g. sudden unexpected death in epilepsy, accidents, drowning) or indirect effects of seizures and therapies. Extensive research with animal models and human studies provides limited insights into the mechanisms underlying seizures and epileptogenesis, and these have not translated into significant reductions in pharmaco-resistance, morbidities or mortality. To help define changes in molecular signalling networks associated with seizures in epilepsy with a broad range of aetiologies, we examined the proteome of brain samples from epilepsy and control cases. Label-free quantitative mass spectrometry was performed on the hippocampal cornu ammonis 1-3 region (CA1-3), frontal cortex and dentate gyrus microdissected from epilepsy and control cases (n = 14/group). Epilepsy cases had significant differences in the expression of 777 proteins in the hippocampal CA1 - 3 region, 296 proteins in the frontal cortex and 49 proteins in the dentate gyrus in comparison to control cases. Network analysis showed that proteins involved in protein synthesis, mitochondrial function, G-protein signalling and synaptic plasticity were particularly altered in epilepsy. While protein differences were most pronounced in the hippocampus, similar changes were observed in other brain regions indicating broad proteomic abnormalities in epilepsy. Among the most significantly altered proteins, G-protein subunit beta 1 (GNB1) was one of the most significantly decreased proteins in epilepsy in all regions studied, highlighting the importance of G-protein subunit signalling and G-protein-coupled receptors in epilepsy. Our results provide insights into common molecular mechanisms underlying epilepsy across various aetiologies, which may allow for novel targeted therapeutic strategies.
PMCID:8214864
PMID: 34159317
ISSN: 2632-1297
CID: 5387022

Metabolic Regulation of the Epigenome Drives Lethal Infantile Ependymoma

Michealraj, Kulandaimanuvel Antony; Kumar, Sachin A; Kim, Leo J Y; Cavalli, Florence M G; Przelicki, David; Wojcik, John B; Delaidelli, Alberto; Bajic, Andrea; Saulnier, Olivier; MacLeod, Graham; Vellanki, Ravi N; Vladoiu, Maria C; Guilhamon, Paul; Ong, Winnie; Lee, John J Y; Jiang, Yanqing; Holgado, Borja L; Rasnitsyn, Alex; Malik, Ahmad A; Tsai, Ricky; Richman, Cory M; Juraschka, Kyle; Haapasalo, Joonas; Wang, Evan Y; De Antonellis, Pasqualino; Suzuki, Hiromichi; Farooq, Hamza; Balin, Polina; Kharas, Kaitlin; Van Ommeren, Randy; Sirbu, Olga; Rastan, Avesta; Krumholtz, Stacey L; Ly, Michelle; Ahmadi, Moloud; Deblois, Geneviève; Srikanthan, Dilakshan; Luu, Betty; Loukides, James; Wu, Xiaochong; Garzia, Livia; Ramaswamy, Vijay; Kanshin, Evgeny; Sánchez-Osuna, María; El-Hamamy, Ibrahim; Coutinho, Fiona J; Prinos, Panagiotis; Singh, Sheila; Donovan, Laura K; Daniels, Craig; Schramek, Daniel; Tyers, Mike; Weiss, Samuel; Stein, Lincoln D; Lupien, Mathieu; Wouters, Bradly G; Garcia, Benjamin A; Arrowsmith, Cheryl H; Sorensen, Poul H; Angers, Stephane; Jabado, Nada; Dirks, Peter B; Mack, Stephen C; Agnihotri, Sameer; Rich, Jeremy N; Taylor, Michael D
Posterior fossa A (PFA) ependymomas are lethal malignancies of the hindbrain in infants and toddlers. Lacking highly recurrent somatic mutations, PFA ependymomas are proposed to be epigenetically driven tumors for which model systems are lacking. Here we demonstrate that PFA ependymomas are maintained under hypoxia, associated with restricted availability of specific metabolites to diminish histone methylation, and increase histone demethylation and acetylation at histone 3 lysine 27 (H3K27). PFA ependymomas initiate from a cell lineage in the first trimester of human development that resides in restricted oxygen. Unlike other ependymomas, transient exposure of PFA cells to ambient oxygen induces irreversible cellular toxicity. PFA tumors exhibit a low basal level of H3K27me3, and, paradoxically, inhibition of H3K27 methylation specifically disrupts PFA tumor growth. Targeting metabolism and/or the epigenome presents a unique opportunity for rational therapy for infants with PFA ependymoma.
PMID: 32445698
ISSN: 1097-4172
CID: 5228932

Mubritinib Targets the Electron Transport Chain Complex I and Reveals the Landscape of OXPHOS Dependency in Acute Myeloid Leukemia

Baccelli, Irène; Gareau, Yves; Lehnertz, Bernhard; Gingras, Stéphane; Spinella, Jean-François; Corneau, Sophie; Mayotte, Nadine; Girard, Simon; Frechette, Mélanie; Blouin-Chagnon, Valérie; Leveillé, Koryne; Boivin, Isabel; MacRae, Tara; Krosl, Jana; Thiollier, Clarisse; Lavallée, Vincent-Philippe; Kanshin, Evgeny; Bertomeu, Thierry; Coulombe-Huntington, Jasmin; St-Denis, Corinne; Bordeleau, Marie-Eve; Boucher, Geneviève; Roux, Philippe P; Lemieux, Sébastien; Tyers, Mike; Thibault, Pierre; Hébert, Josée; Marinier, Anne; Sauvageau, Guy
To identify therapeutic targets in acute myeloid leukemia (AML), we chemically interrogated 200 sequenced primary specimens. Mubritinib, a known ERBB2 inhibitor, elicited strong anti-leukemic effects in vitro and in vivo. In the context of AML, mubritinib functions through ubiquinone-dependent inhibition of electron transport chain (ETC) complex I activity. Resistance to mubritinib characterized normal CD34+ hematopoietic cells and chemotherapy-sensitive AMLs, which displayed transcriptomic hallmarks of hypoxia. Conversely, sensitivity correlated with mitochondrial function-related gene expression levels and characterized a large subset of chemotherapy-resistant AMLs with oxidative phosphorylation (OXPHOS) hyperactivity. Altogether, our work thus identifies an ETC complex I inhibitor and reveals the genetic landscape of OXPHOS dependency in AML.
PMID: 31287994
ISSN: 1878-3686
CID: 5228922

Combined Enrichment/Enzymatic Approach To Study Tightly Clustered Multisite Phosphorylation on Ser-Rich Domains

Kanshin, Evgeny; Pascariu, Mirela; Tyers, Mike; D'Amours, Damien; Thibault, Pierre
The regulation of protein function through phosphorylation is often dominated by allosteric interactions and conformational changes. However, alternative mechanisms involving electrostatic interactions also regulate protein function. In particular, phosphorylation of clusters of Ser/Thr residues can affect protein-plasma membrane/chromatin interactions by electrostatic interactions between phosphosites and phospholipids or histones. Currently, only a few examples of such mechanisms are reported, primarily because of the difficulties of detecting highly phosphorylated proteins and peptides, due in part to the low ionization efficiency and fragmentation yield of multiphosphorylated peptides in mass spectrometry when using positive ion mode detection. This difficulty in detection has resulted in under-reporting of such modified regions, which can be thought of as phosphoproteomic dark matter. Here, we present a novel approach that enriches for multisite-phosphorylated peptides that until now remained inaccessible by conventional phosphoproteomics. Our technique enables the identification of multisite-phosphorylated regions on more than 300 proteins in both yeast and human cells and can be used to profile changes in multisite phosphorylation upon cell stimulation. We further characterize the role of multisite phosphorylation for Ste20 in the yeast mating pheromone response. Mutagenesis experiments confirmed that multisite phosphorylation of Ser/Thr-rich regions plays an important role in the regulation of Ste20 activity during mating pheromone signaling. The ability to detect protein multisite phosphorylation opens new avenues to explore phosphoproteomic dark matter and to study Ser-rich proteins that interact with binding partners through charge pairing mechanisms.
PMID: 30063138
ISSN: 1535-3907
CID: 5228912

Machine Learning of Global Phosphoproteomic Profiles Enables Discrimination of Direct versus Indirect Kinase Substrates

Kanshin, Evgeny; Giguère, Sébastien; Jing, Cheng; Tyers, Mike; Thibault, Pierre
Mass spectrometry allows quantification of tens of thousands of phosphorylation sites from minute amounts of cellular material. Despite this wealth of information, our understanding of phosphorylation-based signaling is limited, in part because it is not possible to deconvolute substrate phosphorylation that is directly mediated by a particular kinase versus phosphorylation that is mediated by downstream kinases. Here, we describe a framework for assignment of direct in vivo kinase substrates using a combination of selective chemical inhibition, quantitative phosphoproteomics, and machine learning techniques. Our workflow allows classification of phosphorylation events following inhibition of an analog-sensitive kinase into kinase-independent effects of the inhibitor, direct effects on cognate substrates, and indirect effects mediated by downstream kinases or phosphatases. We applied this method to identify many direct targets of Cdc28 and Snf1 kinases in the budding yeast Saccharomyces cerevisiae Global phosphoproteome analysis of acute time-series demonstrated that dephosphorylation of direct kinase substrates occurs more rapidly compared with indirect substrates, both after inhibitor treatment and under a physiological nutrient shift in wt cells. Mutagenesis experiments revealed a high proportion of functionally relevant phosphorylation sites on Snf1 targets. For example, Snf1 itself was inhibited through autophosphorylation on Ser391 and new phosphosites were discovered that modulate the activity of the Reg1 regulatory subunit of the Glc7 phosphatase and the Gal83 β-subunit of SNF1 complex. This methodology applies to any kinase for which a functional analog sensitive version can be constructed to facilitate the dissection of the global phosphorylation network.
PMCID:5417821
PMID: 28265048
ISSN: 1535-9484
CID: 5228902

Sample Collection Method Bias Effects in Quantitative Phosphoproteomics

Kanshin, Evgeny; Tyers, Michael; Thibault, Pierre
Current advances in selective enrichment, fractionation, and MS detection of phosphorylated peptides allowed identification and quantitation of tens of thousands phosphosites from minute amounts of biological material. One of the major challenges in the field is preserving the in vivo phosphorylation state of the proteins throughout the sample preparation workflow. This is typically achieved by using phosphatase inhibitors and denaturing conditions during cell lysis. Here we determine if the upstream cell collection techniques could introduce changes in protein phosphorylation. To evaluate the effect of sample collection protocols on the global phosphorylation status of the cell, we compared different sample workflows by metabolic labeling and quantitative mass spectrometry on Saccharomyces cerevisiae cell cultures. We identified highly similar phosphopeptides for cells harvested in ice cold isotonic phosphate buffer, cold ethanol, trichloroacetic acid, and liquid nitrogen. However, quantitative analyses revealed that the commonly used phosphate buffer unexpectedly activated signaling events. Such effects may introduce systematic bias in phosphoproteomics measurements and biochemical analysis.
PMID: 26040406
ISSN: 1535-3907
CID: 5228892