1,653 results on '"Burlingame, Alma L."'
Search Results
2. Phosphorylation-driven epichaperome assembly is a regulator of cellular adaptability and proliferation
- Author
-
Roychowdhury, Tanaya, McNutt, Seth W., Pasala, Chiranjeevi, Nguyen, Hieu T., Thornton, Daniel T., Sharma, Sahil, Botticelli, Luke, Digwal, Chander S., Joshi, Suhasini, Yang, Nan, Panchal, Palak, Chakrabarty, Souparna, Bay, Sadik, Markov, Vladimir, Kwong, Charlene, Lisanti, Jeanine, Chung, Sun Young, Ginsberg, Stephen D., Yan, Pengrong, De Stanchina, Elisa, Corben, Adriana, Modi, Shanu, Alpaugh, Mary L., Colombo, Giorgio, Erdjument-Bromage, Hediye, Neubert, Thomas A., Chalkley, Robert J., Baker, Peter R., Burlingame, Alma L., Rodina, Anna, Chiosis, Gabriela, and Chu, Feixia
- Published
- 2024
- Full Text
- View/download PDF
3. A Legionella toxin exhibits tRNA mimicry and glycosyl transferase activity to target the translation machinery and trigger a ribotoxic stress response
- Author
-
Subramanian, Advait, Wang, Lan, Moss, Tom, Voorhies, Mark, Sangwan, Smriti, Stevenson, Erica, Pulido, Ernst H, Kwok, Samentha, Chalkley, Robert J, Li, Kathy H, Krogan, Nevan J, Swaney, Danielle L, Burlingame, Alma L, Floor, Stephen N, Sil, Anita, Walter, Peter, and Mukherjee, Shaeri
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Lung ,Pneumonia & Influenza ,Infectious Diseases ,Pneumonia ,Genetics ,2.1 Biological and endogenous factors ,2.2 Factors relating to the physical environment ,Infection ,Humans ,Legionella ,Cryoelectron Microscopy ,Legionella pneumophila ,Legionnaires' Disease ,Transferases ,Bacterial Proteins ,Medical and Health Sciences ,Developmental Biology ,Biochemistry and cell biology - Abstract
A widespread strategy employed by pathogens to establish infection is to inhibit host-cell protein synthesis. Legionella pneumophila, an intracellular bacterial pathogen and the causative organism of Legionnaires' disease, secretes a subset of protein effectors into host cells that inhibit translation elongation. Mechanistic insights into how the bacterium targets translation elongation remain poorly defined. We report here that the Legionella effector SidI functions in an unprecedented way as a transfer-RNA mimic that directly binds to and glycosylates the ribosome. The 3.1 Å cryo-electron microscopy structure of SidI reveals an N-terminal domain with an 'inverted L' shape and surface-charge distribution characteristic of tRNA mimicry, and a C-terminal domain that adopts a glycosyl transferase fold that licenses SidI to utilize GDP-mannose as a sugar precursor. This coupling of tRNA mimicry and enzymatic action endows SidI with the ability to block protein synthesis with a potency comparable to ricin, one of the most powerful toxins known. In Legionella-infected cells, the translational pausing activated by SidI elicits a stress response signature mimicking the ribotoxic stress response, which is activated by elongation inhibitors that induce ribosome collisions. SidI-mediated effects on the ribosome activate the stress kinases ZAKα and p38, which in turn drive an accumulation of the protein activating transcription factor 3 (ATF3). Intriguingly, ATF3 escapes the translation block imposed by SidI, translocates to the nucleus and orchestrates the transcription of stress-inducible genes that promote cell death, revealing a major role for ATF3 in the response to collided ribosome stress. Together, our findings elucidate a novel mechanism by which a pathogenic bacterium employs tRNA mimicry to hijack a ribosome-to-nuclear signalling pathway that regulates cell fate.
- Published
- 2023
4. Non-specific recognition of histone modifications by H3K9bhb antibody
- Author
-
Tsusaka, Takeshi, Oses-Prieto, Juan A, Lee, Christina, DeFelice, Brian C, Burlingame, Alma L, and Goldberg, Emily L
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Genetics ,Biotechnology ,Underpinning research ,1.1 Normal biological development and functioning ,Biochemistry ,Biological sciences ,Cell biology ,Molecular biology - Abstract
Ketone bodies are short-chain fatty acids produced in the liver during periods of limited glucose availability that provide an alternative energy source for the brain, heart, and skeletal muscle. Beyond this metabolic role, β-hydroxybutyrate (BHB), is gaining recognition as a signaling molecule. Lysine β-hydroxybutyrylation (Kbhb) is a newly discovered post-translational modification in which BHB is covalently attached to lysine ε-amino groups. This protein adduct is metabolically sensitive, dependent on BHB concentration, and found on proteins in multiple intracellular compartments. Therefore, Kbhb is hypothesized to be an important component of ketone body-regulated physiology. Kbhb on histones is proposed to be an epigenetic regulator, which links metabolic alterations to gene expression. However, we found that the widely used antibody against β-hydroxybutyrylated lysine 9 on histone H3 (H3K9bhb) also recognizes other modification(s) that likely include acetylation. Therefore, caution must be used when interpreting gene regulation data acquired with the H3K9bhb antibody.
- Published
- 2023
5. Chemical Genetic Identification of PKC Epsilon Substrates in Mouse Brain.
- Author
-
Dugan, Michael P, Ferguson, Laura B, Hertz, Nicholas T, Chalkley, Robert J, Burlingame, Alma L, Shokat, Kevan M, Parker, Peter J, and Messing, Robert O
- Subjects
Brain ,Animals ,Mice ,Ethanol ,Alcohol Drinking ,Signal Transduction ,Protein Kinase C-epsilon ,Alcoholism ,Alcohol Use and Health ,Behavioral and Social Science ,Basic Behavioral and Social Science ,Substance Misuse ,Neurosciences ,Genetics ,Brain Disorders ,Aetiology ,2.1 Biological and endogenous factors ,Mental health ,Good Health and Well Being ,Biochemistry & Molecular Biology - Abstract
PKC epsilon (PKCε) plays important roles in behavioral responses to alcohol and in anxiety-like behavior in rodents, making it a potential drug target for reducing alcohol consumption and anxiety. Identifying signals downstream of PKCε could reveal additional targets and strategies for interfering with PKCε signaling. We used a chemical genetic screen combined with mass spectrometry to identify direct substrates of PKCε in mouse brain and validated findings for 39 of them using peptide arrays and in vitro kinase assays. Prioritizing substrates with several public databases such as LINCS-L1000, STRING, GeneFriends, and GeneMAINA predicted interactions between these putative substrates and PKCε and identified substrates associated with alcohol-related behaviors, actions of benzodiazepines, and chronic stress. The 39 substrates could be broadly classified in three functional categories: cytoskeletal regulation, morphogenesis, and synaptic function. These results provide a list of brain PKCε substrates, many of which are novel, for future investigation to determine the role of PKCε signaling in alcohol responses, anxiety, responses to stress, and other related behaviors.
- Published
- 2023
6. Capture, Release, and Identification of Newly Synthesized Proteins for Improved Profiling of Functional Translatomes
- Author
-
Phillips, Nancy J, Vinaithirthan, Bala M, Oses-Prieto, Juan A, Chalkley, Robert J, and Burlingame, Alma L
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Genetics ,Biotin ,Azides ,Proteins ,Peptides ,RNA ,Messenger ,O-propargyl-puromycin ,cleavable linker ,mTOR ,nascent proteins ,translation ,Biochemistry & Molecular Biology - Abstract
New protein synthesis is regulated both at the level of mRNA transcription and translation. RNA-Seq is effective at measuring levels of mRNA expression, but techniques to monitor mRNA translation are much more limited. Previously, we reported results from O-propargyl-puromycin (OPP) labeling of proteins undergoing active translation in a 2-h time frame, followed by biotinylation using click chemistry, affinity purification, and on-bead digestion to identify nascent proteins by mass spectrometry (OPP-ID). As with any on-bead digestion protocol, the problem of nonspecific binders complicated the rigorous categorization of nascent proteins by OPP-ID. Here, we incorporate a chemically cleavable linker, Dde biotin-azide, into the protocol (OPP-IDCL) to provide specific release of modified proteins from the streptavidin beads. Following capture, the Dde moiety is readily cleaved with 2% hydrazine, releasing nascent polypeptides bearing OPP plus a residual C3H8N4 tag. When results are compared side by side with the original OPP-ID method, change to a cleavable linker led to a dramatic reduction in the number of background proteins detected in controls and a concomitant increase in the number of proteins that could be characterized as newly synthesized. We evaluated the method's ability to detect nascent proteins at various submilligram protein input levels and showed that, when starting with only 100 μg of protein, ∼1500 nascent proteins could be identified with low background. Upon treatment of K562 cells with MLN128, a potent inhibitor of the mammalian target of rapamycin, prior to OPP treatment, we identified 1915 nascent proteins, the majority of which were downregulated upon inhibitor treatment. Repressed proteins with log2 FC
- Published
- 2023
7. Reversible lysine-targeted probes reveal residence time-based kinase selectivity
- Author
-
Yang, Tangpo, Cuesta, Adolfo, Wan, Xiaobo, Craven, Gregory B, Hirakawa, Brad, Khamphavong, Penney, May, Jeffrey R, Kath, John C, Lapek, John D, Niessen, Sherry, Burlingame, Alma L, Carelli, Jordan D, and Taunton, Jack
- Subjects
Biochemistry and Cell Biology ,Chemical Sciences ,Biological Sciences ,Brain Disorders ,Animals ,Cysteine ,Lysine ,Mice ,Protein Binding ,Protein Kinase Inhibitors ,Protein Kinases ,Medicinal and Biomolecular Chemistry ,Biochemistry & Molecular Biology ,Biochemistry and cell biology ,Medicinal and biomolecular chemistry - Abstract
The expansion of the target landscape of covalent inhibitors requires the engagement of nucleophiles beyond cysteine. Although the conserved catalytic lysine in protein kinases is an attractive candidate for a covalent approach, selectivity remains an obvious challenge. Moreover, few covalent inhibitors have been shown to engage the kinase catalytic lysine in animals. We hypothesized that reversible, lysine-targeted inhibitors could provide sustained kinase engagement in vivo, with selectivity driven in part by differences in residence time. By strategically linking benzaldehydes to a promiscuous kinase binding scaffold, we developed chemoproteomic probes that reversibly and covalently engage >200 protein kinases in cells and mice. Probe-kinase residence time was dramatically enhanced by a hydroxyl group ortho to the aldehyde. Remarkably, only a few kinases, including Aurora A, showed sustained, quasi-irreversible occupancy in vivo, the structural basis for which was revealed by X-ray crystallography. We anticipate broad application of salicylaldehyde-based probes to proteins that lack a druggable cysteine.
- Published
- 2022
8. RAPIDASH: Tag-free enrichment of ribosome-associated proteins reveals composition dynamics in embryonic tissue, cancer cells, and macrophages
- Author
-
Susanto, Teodorus Theo, Hung, Victoria, Levine, Andrew G., Chen, Yuxiang, Kerr, Craig H., Yoo, Yongjin, Oses-Prieto, Juan A., Fromm, Lisa, Zhang, Zijian, Lantz, Travis C., Fujii, Kotaro, Wernig, Marius, Burlingame, Alma L., Ruggero, Davide, and Barna, Maria
- Published
- 2024
- Full Text
- View/download PDF
9. Intra-axonal translation of Khsrp mRNA slows axon regeneration by destabilizing localized mRNAs
- Author
-
Patel, Priyanka, Buchanan, Courtney N, Zdradzinski, Matthew D, Sahoo, Pabitra K, Kar, Amar N, Lee, Seung Joon, Vaughn, Lauren S, Urisman, Anatoly, Oses-Prieto, Juan, Dell’Orco, Michela, Cassidy, Devon E, Costa, Irene Dalla, Miller, Sharmina, Thames, Elizabeth, Smith, Terika P, Burlingame, Alma L, Perrone-Bizzozero, Nora, and Twiss, Jeffery L
- Subjects
Genetics ,Peripheral Neuropathy ,Biotechnology ,Neurodegenerative ,Physical Injury - Accidents and Adverse Effects ,Neurosciences ,Regenerative Medicine ,1.1 Normal biological development and functioning ,Underpinning research ,Neurological ,3' Untranslated Regions ,Animals ,Axons ,Mice ,Nerve Regeneration ,RNA ,Messenger ,RNA-Binding Proteins ,Rats ,Rats ,Sprague-Dawley ,Sciatic Nerve ,Environmental Sciences ,Biological Sciences ,Information and Computing Sciences ,Developmental Biology - Abstract
Axonally synthesized proteins support nerve regeneration through retrograde signaling and local growth mechanisms. RNA binding proteins (RBP) are needed for this and other aspects of post-transcriptional regulation of neuronal mRNAs, but only a limited number of axonal RBPs are known. We used targeted proteomics to profile RBPs in peripheral nerve axons. We detected 76 proteins with reported RNA binding activity in axoplasm, and levels of several change with axon injury and regeneration. RBPs with altered levels include KHSRP that decreases neurite outgrowth in developing CNS neurons. Axonal KHSRP levels rapidly increase after injury remaining elevated up to 28 days post axotomy. Khsrp mRNA localizes into axons and the rapid increase in axonal KHSRP is through local translation of Khsrp mRNA in axons. KHSRP can bind to mRNAs with 3'UTR AU-rich elements and targets those transcripts to the cytoplasmic exosome for degradation. KHSRP knockout mice show increased axonal levels of KHSRP target mRNAs, Gap43, Snap25, and Fubp1, following sciatic nerve injury and these mice show accelerated nerve regeneration in vivo. Together, our data indicate that axonal translation of the RNA binding protein Khsrp mRNA following nerve injury serves to promote decay of other axonal mRNAs and slow axon regeneration.
- Published
- 2022
10. Covalent labeling of a chromatin reader domain using proximity-reactive cyclic peptides
- Author
-
Zhang, Meng Yao, Yang, Hyunjun, Ortiz, Gloria, Trnka, Michael J, Petronikolou, Nektaria, Burlingame, Alma L, DeGrado, William F, and Fujimori, Danica Galonić
- Subjects
Organic Chemistry ,Chemical Sciences ,Genetics ,Biotechnology ,Chemical sciences - Abstract
Chemical probes for chromatin reader proteins are valuable tools for investigating epigenetic regulatory mechanisms and evaluating whether the target of interest holds therapeutic potential. Developing potent inhibitors for the plant homeodomain (PHD) family of methylation readers remains a difficult task due to the charged, shallow and extended nature of the histone binding site that precludes effective engagement of conventional small molecules. Herein, we describe the development of novel proximity-reactive cyclopeptide inhibitors for PHD3-a trimethyllysine reader domain of histone demethylase KDM5A. Guided by the PHD3-histone co-crystal structure, we designed a sidechain-to-sidechain linking strategy to improve peptide proteolytic stability whilst maintaining binding affinity. We have developed an operationally simple solid-phase macrocyclization pathway, capitalizing on the inherent reactivity of the dimethyllysine ε-amino group to generate scaffolds bearing charged tetraalkylammonium functionalities that effectively engage the shallow aromatic 'groove' of PHD3. Leveraging a surface-exposed lysine residue on PHD3 adjacent to the ligand binding site, cyclic peptides were rendered covalent through installation of an arylsulfonyl fluoride warhead. The resulting lysine-reactive cyclic peptides demonstrated rapid and efficient labeling of the PHD3 domain in HEK293T lysates, showcasing the feasibility of employing proximity-induced reactivity for covalent labeling of this challenging family of reader domains.
- Published
- 2022
11. Hemin-Induced Death Models Hemorrhagic Stroke and Is a Variant of Classical Neuronal Ferroptosis.
- Author
-
Zille, Marietta, Oses-Prieto, Juan A, Savage, Sara R, Karuppagounder, Saravanan S, Chen, Yingxin, Kumar, Amit, Morris, John H, Scheidt, Karl A, Burlingame, Alma L, and Ratan, Rajiv R
- Subjects
Biomedical and Clinical Sciences ,Neurosciences ,Stroke ,Hematology ,Brain Disorders ,2.1 Biological and endogenous factors ,Aetiology ,Neurological ,Animals ,Antioxidants ,Extracellular Signal-Regulated MAP Kinases ,Ferroptosis ,Glutathione ,Hemin ,Hemoglobins ,Hemorrhagic Stroke ,Intracranial Hemorrhages ,Iron ,Male ,Mice ,Mitogen-Activated Protein Kinase Kinases ,Necrosis ,Neurons ,Phosphorylation ,brain hemorrhage ,cell death ,ferroptosis ,MAP signaling ,phosphoproteomics ,stroke ,Medical and Health Sciences ,Psychology and Cognitive Sciences ,Neurology & Neurosurgery - Abstract
Ferroptosis is a caspase-independent, iron-dependent form of regulated necrosis extant in traumatic brain injury, Huntington disease, and hemorrhagic stroke. It can be activated by cystine deprivation leading to glutathione depletion, the insufficiency of the antioxidant glutathione peroxidase-4, and the hemolysis products hemoglobin and hemin. A cardinal feature of ferroptosis is extracellular signal-regulated kinase (ERK)1/2 activation culminating in its translocation to the nucleus. We have previously confirmed that the mitogen-activated protein (MAP) kinase kinase (MEK) inhibitor U0126 inhibits persistent ERK1/2 phosphorylation and ferroptosis. Here, we show that hemin exposure, a model of secondary injury in brain hemorrhage and ferroptosis, activated ERK1/2 in mouse neurons. Accordingly, MEK inhibitor U0126 protected against hemin-induced ferroptosis. Unexpectedly, U0126 prevented hemin-induced ferroptosis independent of its ability to inhibit ERK1/2 signaling. In contrast to classical ferroptosis in neurons or cancer cells, chemically diverse inhibitors of MEK did not block hemin-induced ferroptosis, nor did the forced expression of the ERK-selective MAP kinase phosphatase (MKP)3. We conclude that hemin or hemoglobin-induced ferroptosis, unlike glutathione depletion, is ERK1/2-independent. Together with recent studies, our findings suggest the existence of a novel subtype of neuronal ferroptosis relevant to bleeding in the brain that is 5-lipoxygenase-dependent, ERK-independent, and transcription-independent. Remarkably, our unbiased phosphoproteome analysis revealed dramatic differences in phosphorylation induced by two ferroptosis subtypes. As U0126 also reduced cell death and improved functional recovery after hemorrhagic stroke in male mice, our analysis also provides a template on which to build a search for U0126's effects in a variant of neuronal ferroptosis.SIGNIFICANCE STATEMENT Ferroptosis is an iron-dependent mechanism of regulated necrosis that has been linked to hemorrhagic stroke. Common features of ferroptotic death induced by diverse stimuli are the depletion of the antioxidant glutathione, production of lipoxygenase-dependent reactive lipids, sensitivity to iron chelation, and persistent activation of extracellular signal-regulated kinase (ERK) signaling. Unlike classical ferroptosis induced in neurons or cancer cells, here we show that ferroptosis induced by hemin is ERK-independent. Paradoxically, the canonical MAP kinase kinase (MEK) inhibitor U0126 blocks brain hemorrhage-induced death. Altogether, these data suggest that a variant of ferroptosis is unleashed in hemorrhagic stroke. We present the first, unbiased phosphoproteomic analysis of ferroptosis as a template on which to understand distinct paths to cell death that meet the definition of ferroptosis.
- Published
- 2022
12. Modulating environmental signals to reveal mechanisms and vulnerabilities of cancer persisters
- Author
-
Sun, Xiaoxiao, Bieber, Jake M, Hammerlindl, Heinz, Chalkley, Robert J, Li, Kathy H, Burlingame, Alma L, Jacobson, Matthew P, Wu, Lani F, and Altschuler, Steven J
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Cancer ,Development of treatments and therapeutic interventions ,5.1 Pharmaceuticals ,Good Health and Well Being ,Anti-Bacterial Agents ,Humans ,Neoplasms - Abstract
Cancer persister cells are able to survive otherwise lethal doses of drugs through nongenetic mechanisms, which can lead to cancer regrowth and drug resistance. The broad spectrum of molecular differences observed between persisters and their treatment-naïve counterparts makes it challenging to identify causal mechanisms underlying persistence. Here, we modulate environmental signals to identify cellular mechanisms that promote the emergence of persisters and to pinpoint actionable vulnerabilities that eliminate them. We found that interferon-γ (IFNγ) can induce a pro-persistence signal that can be specifically eliminated by inhibition of type I protein arginine methyltransferase (PRMT) (PRMTi). Mechanistic investigation revealed that signal transducer and activator of transcription 1 (STAT1) is a key component connecting IFNγ's pro-persistence and PRMTi's antipersistence effects, suggesting a previously unknown application of PRMTi to target persisters in settings with high STAT1 expression. Modulating environmental signals can accelerate the identification of mechanisms that promote and eliminate cancer persistence.
- Published
- 2022
13. A stem cell roadmap of ribosome heterogeneity reveals a function for RPL10A in mesoderm production
- Author
-
Genuth, Naomi R, Shi, Zhen, Kunimoto, Koshi, Hung, Victoria, Xu, Adele F, Kerr, Craig H, Tiu, Gerald C, Oses-Prieto, Juan A, Salomon-Shulman, Rachel EA, Axelrod, Jeffrey D, Burlingame, Alma L, Loh, Kyle M, and Barna, Maria
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Genetics ,Stem Cell Research ,Stem Cell Research - Nonembryonic - Non-Human ,Underpinning research ,1.1 Normal biological development and functioning ,Generic health relevance ,Animals ,Cell Differentiation ,Humans ,Mesoderm ,Mice ,Ribosomal Proteins ,Ribosomes ,Stem Cells ,Wnt Signaling Pathway - Abstract
Recent findings suggest that the ribosome itself modulates gene expression. However, whether ribosomes change composition across cell types or control cell fate remains unknown. Here, employing quantitative mass spectrometry during human embryonic stem cell differentiation, we identify dozens of ribosome composition changes underlying cell fate specification. We observe upregulation of RPL10A/uL1-containing ribosomes in the primitive streak followed by progressive decreases during mesoderm differentiation. An Rpl10a loss-of-function allele in mice causes striking early mesodermal phenotypes, including posterior trunk truncations, and inhibits paraxial mesoderm production in culture. Ribosome profiling in Rpl10a loss-of-function mice reveals decreased translation of mesoderm regulators, including Wnt pathway mRNAs, which are also enriched on RPL10A/uL1-containing ribosomes. We further show that RPL10A/uL1 regulates canonical and non-canonical Wnt signaling during stem cell differentiation and in the developing embryo. These findings reveal unexpected ribosome composition modularity that controls differentiation and development through the specialized translation of key signaling networks.
- Published
- 2022
14. TSC1 loss increases risk for tauopathy by inducing tau acetylation and preventing tau clearance via chaperone-mediated autophagy
- Author
-
Alquezar, Carolina, Schoch, Kathleen M, Geier, Ethan G, Ramos, Eliana Marisa, Scrivo, Aurora, Li, Kathy H, Argouarch, Andrea R, Mlynarski, Elisabeth E, Dombroski, Beth, DeTure, Michael, Dickson, Dennis W, Yokoyama, Jennifer S, Cuervo, Ana M, Burlingame, Alma L, Schellenberg, Gerard D, Miller, Timothy M, Miller, Bruce L, and Kao, Aimee W
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Dementia ,Alzheimer's Disease ,Rare Diseases ,Alzheimer's Disease Related Dementias (ADRD) ,Frontotemporal Dementia (FTD) ,Neurosciences ,Neurodegenerative ,Aging ,Acquired Cognitive Impairment ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Genetics ,Brain Disorders ,2.1 Biological and endogenous factors - Abstract
Age-associated neurodegenerative disorders demonstrating tau-laden intracellular inclusions are known as tauopathies. We previously linked a loss-of-function mutation in the TSC1 gene to tau accumulation and frontotemporal lobar degeneration. Now, we have identified genetic variants in TSC1 that decrease TSC1/hamartin levels and predispose to tauopathies such as Alzheimer’s disease and progressive supranuclear palsy. Cellular and murine models of TSC1 haploinsufficiency, as well as human brains carrying a TSC1 risk variant, accumulated tau protein that exhibited aberrant acetylation. This acetylation hindered tau degradation via chaperone-mediated autophagy, thereby leading to its accumulation. Aberrant tau acetylation in TSC1 haploinsufficiency resulted from the dysregulation of both p300 acetyltransferase and SIRT1 deacetylase. Pharmacological modulation of either enzyme restored tau levels. This study substantiates TSC1 as a novel tauopathy risk gene and includes TSC1 haploinsufficiency as a genetic model for tauopathies. In addition, these findings promote tau acetylation as a rational target for tauopathy therapeutics and diagnostic.
- Published
- 2021
15. Neutrophil-inflicted vasculature damage suppresses immune-mediated optic nerve regeneration
- Author
-
Passino, Ryan, Finneran, Matthew C., Hafner, Hannah, Feng, Qian, Huffman, Lucas D., Zhao, Xiao-Feng, Johnson, Craig N., Kawaguchi, Riki, Oses-Prieto, Juan A., Burlingame, Alma L., Geschwind, Daniel H., Benowitz, Larry I., and Giger, Roman J.
- Published
- 2024
- Full Text
- View/download PDF
16. Antibody-directed extracellular proximity biotinylation reveals that Contactin-1 regulates axo-axonic innervation of axon initial segments
- Author
-
Ogawa, Yuki, Lim, Brian C., George, Shanu, Oses-Prieto, Juan A., Rasband, Joshua M., Eshed-Eisenbach, Yael, Hamdan, Hamdan, Nair, Supna, Boato, Francesco, Peles, Elior, Burlingame, Alma L., Van Aelst, Linda, and Rasband, Matthew N.
- Published
- 2023
- Full Text
- View/download PDF
17. ERα is an RNA-binding protein sustaining tumor cell survival and drug resistance
- Author
-
Xu, Yichen, Huangyang, Peiwei, Wang, Ying, Xue, Lingru, Devericks, Emily, Nguyen, Hao G, Yu, Xiuyan, Oses-Prieto, Juan A, Burlingame, Alma L, Miglani, Sohit, Goodarzi, Hani, and Ruggero, Davide
- Subjects
Genetics ,Cancer ,Breast Cancer ,Human Genome ,Generic health relevance ,Animals ,Base Sequence ,Breast Neoplasms ,Cell Line ,Tumor ,Cell Survival ,Disease Progression ,Drug Resistance ,Neoplasm ,Estrogen Receptor alpha ,Eukaryotic Initiation Factor-4G ,Female ,Gene Expression Regulation ,Neoplastic ,Genomics ,Humans ,Mice ,Inbred NOD ,Myeloid Cell Leukemia Sequence 1 Protein ,Oncogenes ,Protein Binding ,Protein Domains ,RNA Splicing ,RNA ,Messenger ,RNA-Binding Proteins ,Stress ,Physiological ,Tamoxifen ,X-Box Binding Protein 1 ,ERα ,RNA splicing ,RNA-binding protein ,breast cancer ,cell survival ,integrated stress response ,translation control ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology - Abstract
Estrogen receptor α (ERα) is a hormone receptor and key driver for over 70% of breast cancers that has been studied for decades as a transcription factor. Unexpectedly, we discover that ERα is a potent non-canonical RNA-binding protein. We show that ERα RNA binding function is uncoupled from its activity to bind DNA and critical for breast cancer progression. Employing genome-wide cross-linking immunoprecipitation (CLIP) sequencing and a functional CRISPRi screen, we find that ERα-associated mRNAs sustain cancer cell fitness and elicit cellular responses to stress. Mechanistically, ERα controls different steps of RNA metabolism. In particular, we demonstrate that ERα RNA binding mediates alternative splicing of XBP1 and translation of the eIF4G2 and MCL1 mRNAs, which facilitates survival upon stress conditions and sustains tamoxifen resistance of cancer cells. ERα is therefore a multifaceted RNA-binding protein, and this activity transforms our knowledge of post-transcriptional regulation underlying cancer development and drug response.
- Published
- 2021
18. Cell-cell adhesion regulates Merlin/NF2 interaction with the PAF complex
- Author
-
Roehrig, Anne E, Klupsch, Kristina, Oses-Prieto, Juan A, Chaib, Selim, Henderson, Stephen, Emmett, Warren, Young, Lucy C, Surinova, Silvia, Blees, Andreas, Pfeiffer, Anett, Tijani, Maha, Brunk, Fabian, Hartig, Nicole, Muñoz-Alegre, Marta, Hergovich, Alexander, Jennings, Barbara H, Burlingame, Alma L, and Rodriguez-Viciana, Pablo
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Genetics ,Rare Diseases ,Neurofibromatosis ,Neurosciences ,2.1 Biological and endogenous factors ,1.1 Normal biological development and functioning ,Aetiology ,Underpinning research ,Cancer ,Cell Adhesion ,Cell Proliferation ,Chromatin ,Chromatin Assembly and Disassembly ,Contact Inhibition ,DNA Helicases ,DNA-Binding Proteins ,Gene Expression Regulation ,HEK293 Cells ,Humans ,Neoplasms ,Neurofibromin 2 ,Protein Binding ,Protein Interaction Maps ,Signal Transduction ,Tumor Suppressor Proteins ,General Science & Technology - Abstract
The PAF complex (PAFC) coordinates transcription elongation and mRNA processing and its CDC73/parafibromin subunit functions as a tumour suppressor. The NF2/Merlin tumour suppressor functions both at the cell cortex and nucleus and is a key mediator of contact inhibition but the molecular mechanisms remain unclear. In this study we have used affinity proteomics to identify novel Merlin interacting proteins and show that Merlin forms a complex with multiple proteins involved in RNA processing including the PAFC and the CHD1 chromatin remodeller. Tumour-derived inactivating mutations in both Merlin and the CDC73 PAFC subunit mutually disrupt their interaction and growth suppression by Merlin requires CDC73. Merlin interacts with the PAFC in a cell density-dependent manner and we identify a role for FAT cadherins in regulating the Merlin-PAFC interaction. Our results suggest that in addition to its function within the Hippo pathway, Merlin is part of a tumour suppressor network regulated by cell-cell adhesion which coordinates post-initiation steps of the transcription cycle of genes mediating contact inhibition.
- Published
- 2021
19. Age-related loss of neural stem cell O-GlcNAc promotes a glial fate switch through STAT3 activation
- Author
-
White, Charles W, Fan, Xuelai, Maynard, Jason C, Wheatley, Elizabeth G, Bieri, Gregor, Couthouis, Julien, Burlingame, Alma L, and Villeda, Saul A
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Aging ,Brain Disorders ,Regenerative Medicine ,Stem Cell Research ,Stem Cell Research - Nonembryonic - Non-Human ,Neurosciences ,Genetics ,Human Genome ,1.1 Normal biological development and functioning ,Underpinning research ,Neurological ,Animals ,Cell Differentiation ,Cell Proliferation ,Computational Biology ,Gene Expression Regulation ,Glucosamine ,Hippocampus ,Mice ,Neural Stem Cells ,Neurogenesis ,Neuroglia ,STAT3 Transcription Factor ,Sequence Analysis ,RNA ,neural stem cells ,aging ,O-GlcNAcylation ,neurogenesis ,gliogenesis - Abstract
Increased neural stem cell (NSC) quiescence is a major determinant of age-related regenerative decline in the adult hippocampus. However, a coextensive model has been proposed in which division-coupled conversion of NSCs into differentiated astrocytes restrict the stem cell pool with age. Here we report that age-related loss of the posttranslational modification, O-linked β-N-acetylglucosamine (O-GlcNAc), in NSCs promotes a glial fate switch. We detect an age-dependent decrease in NSC O-GlcNAc levels coincident with decreased neurogenesis and increased gliogenesis in the mature hippocampus. Mimicking an age-related loss of NSC O-GlcNAcylation in young mice reduces neurogenesis, increases astrocyte differentiation, and impairs associated cognitive function. Using RNA-sequencing of primary NSCs following decreased O-GlcNAcylation, we detected changes in the STAT3 signaling pathway indicative of glial differentiation. Moreover, using O-GlcNAc-specific mass spectrometry analysis of the aging hippocampus, together with an in vitro site-directed mutagenesis approach, we identify loss of STAT3 O-GlcNAc at Threonine 717 as a driver of astrocyte differentiation. Our data identify the posttranslational modification, O-GlcNAc, as a key molecular regulator of regenerative decline underlying an age-related NSC fate switch.
- Published
- 2020
20. LEM2 phase separation promotes ESCRT-mediated nuclear envelope reformation
- Author
-
von Appen, Alexander, LaJoie, Dollie, Johnson, Isabel E, Trnka, Michael J, Pick, Sarah M, Burlingame, Alma L, Ullman, Katharine S, and Frost, Adam
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Genetics ,1.1 Normal biological development and functioning ,Generic health relevance ,Anaphase ,Chromatin ,DNA Damage ,DNA-Binding Proteins ,Endosomal Sorting Complexes Required for Transport ,HeLa Cells ,Humans ,Membrane Proteins ,Microtubules ,Nuclear Envelope ,Nuclear Proteins ,Spindle Apparatus ,Hela Cells ,General Science & Technology - Abstract
During cell division, remodelling of the nuclear envelope enables chromosome segregation by the mitotic spindle1. The reformation of sealed nuclei requires ESCRTs (endosomal sorting complexes required for transport) and LEM2, a transmembrane ESCRT adaptor2-4. Here we show how the ability of LEM2 to condense on microtubules governs the activation of ESCRTs and coordinated spindle disassembly. The LEM motif of LEM2 binds BAF, conferring on LEM2 an affinity for chromatin5,6, while an adjacent low-complexity domain (LCD) promotes LEM2 phase separation. A proline-arginine-rich sequence within the LCD binds to microtubules and targets condensation of LEM2 to spindle microtubules that traverse the nascent nuclear envelope. Furthermore, the winged-helix domain of LEM2 activates the ESCRT-II/ESCRT-III hybrid protein CHMP7 to form co-oligomeric rings. Disruption of these events in human cells prevented the recruitment of downstream ESCRTs, compromised spindle disassembly, and led to defects in nuclear integrity and DNA damage. We propose that during nuclear reassembly LEM2 condenses into a liquid-like phase and coassembles with CHMP7 to form a macromolecular O-ring seal at the confluence between membranes, chromatin and the spindle. The properties of LEM2 described here, and the homologous architectures of related inner nuclear membrane proteins7,8, suggest that phase separation may contribute to other critical envelope functions, including interphase repair8-13 and chromatin organization14-17.
- Published
- 2020
21. Characterization of Prenylated C-terminal Peptides Using a Thiopropyl-based Capture Technique and LC-MS/MS.
- Author
-
Wilkins, James A, Kaasik, Krista, Chalkley, Robert J, and Burlingame, Alma L
- Subjects
Brain ,Animals ,Mice ,Peptide Hydrolases ,Sepharose ,Peptides ,Proteins ,Chromatography ,Liquid ,Proteomics ,Amino Acid Sequence ,Databases ,Protein ,Tandem Mass Spectrometry ,Protein Prenylation ,Affinity Proteomics ,Cancer Biology ,Cell Biology ,Cellular Organelles ,Cytoskeleton ,Growth Regulation ,Isoprenes ,Nuclear Structure ,Posttranslational Modifications ,Prenylation ,Biotechnology ,Biochemistry & Molecular Biology - Abstract
Posttranslational modifications play a critical and diverse role in regulating cellular activities. Despite their fundamentally important role in cellular function, there has been no report to date of an effective generalized approach to the targeting, extraction, and characterization of the critical c-terminal regions of natively prenylated proteins. Various chemical modification and metabolic labeling strategies in cell culture have been reported. However, their applicability is limited to cell culture systems and does not allow for analysis of tissue samples. The chemical characteristics (hydrophobicity, low abundance, highly basic charge) of many of the c-terminal regions of prenylated proteins have impaired the use of standard proteomic workflows. In this context, we sought a direct approach to the problem in order to examine these proteins in tissue without the use of labeling. Here we demonstrate that prenylated proteins can be captured on chromatographic resins functionalized with mixed disulfide functions. Protease treatment of resin-bound proteins using chymotryptic digestion revealed peptides from many known prenylated proteins. Exposure of the protease-treated resin to reducing agents and hydro organic mixtures released c-terminal peptides with intact prenyl groups along with other enzymatic modifications expected in this protein family. Database and search parameters were selected to allow for c-terminal modifications unique to these molecules such as CAAX box processing and c-terminal methylation. In summary, we present a direct approach to enrich and obtain information at a molecular level of detail about prenylation of proteins from tissue and cell extracts using high-performance LC-MS without the need for metabolic labeling and derivatization.
- Published
- 2020
22. NuMA1 promotes axon initial segment assembly through inhibition of endocytosis
- Author
-
Torii, Tomohiro, Ogawa, Yuki, Liu, Cheng-Hsin, Ho, Tammy Szu-Yu, Hamdan, Hamdan, Wang, Chih-chuan, Oses-Prieto, Juan A, Burlingame, Alma L, and Rasband, Matthew N
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Neurosciences ,Brain Disorders ,Pediatric ,Underpinning research ,1.1 Normal biological development and functioning ,1-Alkyl-2-acetylglycerophosphocholine Esterase ,Animals ,Ankyrins ,Axon Initial Segment ,Axons ,Cell Adhesion Molecules ,Cell Cycle Proteins ,Cytoskeleton ,Dyneins ,Endocytosis ,Gene Expression Regulation ,Humans ,Mice ,Microfilament Proteins ,Microtubule-Associated Proteins ,Nerve Growth Factors ,Neurons ,Protein Transport ,Proteomics ,RNA ,Small Interfering ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
Axon initial segments (AISs) initiate action potentials and regulate the trafficking of vesicles between somatodendritic and axonal compartments. However, the mechanisms controlling AIS assembly remain poorly defined. We performed differential proteomics and found nuclear mitotic apparatus protein 1 (NuMA1) is downregulated in AIS-deficient neonatal mouse brains and neurons. NuMA1 is transiently located at the AIS during development where it interacts with the scaffolding protein 4.1B and the dynein regulator lissencephaly 1 (Lis1). Silencing NuMA1 or protein 4.1B by shRNA disrupts AIS assembly, but not maintenance. Silencing Lis1 or overexpressing NuMA1 during AIS assembly increased the density of AIS proteins, including ankyrinG and neurofascin-186 (NF186). NuMA1 inhibits the endocytosis of AIS NF186 by impeding Lis1's interaction with doublecortin, a potent facilitator of NF186 endocytosis. Our results indicate the transient expression and AIS localization of NuMA1 stabilizes the developing AIS by inhibiting endocytosis and removal of AIS proteins.
- Published
- 2020
23. Mapping axon initial segment structure and function by multiplexed proximity biotinylation.
- Author
-
Hamdan, Hamdan, Lim, Brian C, Torii, Tomohiro, Joshi, Abhijeet, Konning, Matthias, Smith, Cameron, Palmer, Donna J, Ng, Philip, Leterrier, Christophe, Oses-Prieto, Juan A, Burlingame, Alma L, and Rasband, Matthew N
- Subjects
Neurons ,Axons ,Animals ,Humans ,Mice ,Rats ,Rats ,Sprague-Dawley ,Proteome ,Biotinylation ,Protein Transport ,Mass Spectrometry ,Axon Initial Segment ,Sprague-Dawley - Abstract
Axon initial segments (AISs) generate action potentials and regulate the polarized distribution of proteins, lipids, and organelles in neurons. While the mechanisms of AIS Na+ and K+ channel clustering are understood, the molecular mechanisms that stabilize the AIS and control neuronal polarity remain obscure. Here, we use proximity biotinylation and mass spectrometry to identify the AIS proteome. We target the biotin-ligase BirA* to the AIS by generating fusion proteins of BirA* with NF186, Ndel1, and Trim46; these chimeras map the molecular organization of AIS intracellular membrane, cytosolic, and microtubule compartments. Our experiments reveal a diverse set of biotinylated proteins not previously reported at the AIS. We show many are located at the AIS, interact with known AIS proteins, and their loss disrupts AIS structure and function. Our results provide conceptual insights and a resource for AIS molecular organization, the mechanisms of AIS stability, and polarized trafficking in neurons.
- Published
- 2020
24. An ER translocon for multi-pass membrane protein biogenesis
- Author
-
McGilvray, Philip T, Anghel, S Andre, Sundaram, Arunkumar, Zhong, Frank, Trnka, Michael J, Fuller, James R, Hu, Hong, Burlingame, Alma L, and Keenan, Robert J
- Subjects
Underpinning research ,1.1 Normal biological development and functioning ,Generic health relevance ,Cell Line ,Cryoelectron Microscopy ,Endoplasmic Reticulum ,Humans ,Membrane Proteins ,Protein Biosynthesis ,Protein Domains ,Ribosomes ,SEC Translocation Channels ,biochemistry ,biogenesis ,cell biology ,chemical biology ,co-translational ,endoplasmic reticulum ,folding ,human ,human disease ,insertion ,multi-pass membrane protein ,ribosome ,translocon ,Biochemistry and Cell Biology - Abstract
Membrane proteins with multiple transmembrane domains play critical roles in cell physiology, but little is known about the machinery coordinating their biogenesis at the endoplasmic reticulum. Here we describe a ~ 360 kDa ribosome-associated complex comprising the core Sec61 channel and five accessory factors: TMCO1, CCDC47 and the Nicalin-TMEM147-NOMO complex. Cryo-electron microscopy reveals a large assembly at the ribosome exit tunnel organized around a central membrane cavity. Similar to protein-conducting channels that facilitate movement of transmembrane segments, cytosolic and luminal funnels in TMCO1 and TMEM147, respectively, suggest routes into the central membrane cavity. High-throughput mRNA sequencing shows selective translocon engagement with hundreds of different multi-pass membrane proteins. Consistent with a role in multi-pass membrane protein biogenesis, cells lacking different accessory components show reduced levels of one such client, the glutamate transporter EAAT1. These results identify a new human translocon and provide a molecular framework for understanding its role in multi-pass membrane protein biogenesis.
- Published
- 2020
25. Mechanisms underlying the synaptic trafficking of the glutamate delta receptor GluD1
- Author
-
Tao, Wucheng, Ma, Chenxue, Bemben, Michael A, Li, Kathy H, Burlingame, Alma L, Zhang, Mingjie, and Nicoll, Roger A
- Subjects
Pharmacology and Pharmaceutical Sciences ,Biomedical and Clinical Sciences ,Schizophrenia ,Mental Health ,Neurosciences ,Brain Disorders ,Animals ,Carrier Proteins ,Glutamate Dehydrogenase ,Glutamic Acid ,Hippocampus ,Long-Term Potentiation ,Membrane Proteins ,Mice ,Neuronal Plasticity ,Neurons ,Patch-Clamp Techniques ,Protein Binding ,Protein Transport ,Receptors ,AMPA ,Receptors ,Glutamate ,Receptors ,Opioid ,delta ,Synapses ,Biological Sciences ,Medical and Health Sciences ,Psychology and Cognitive Sciences ,Psychiatry ,Clinical sciences ,Biological psychology ,Clinical and health psychology - Abstract
Ionotropic glutamate delta receptors do not bind glutamate and do not generate ionic current, resulting in difficulty in studying the function and trafficking of these receptors. Here, we utilize chimeric constructs, in which the ligand-binding domain of GluD1 is replaced by that of GluK1, to examine its synaptic trafficking and plasticity. GluD1 trafficked to the synapse, but was incapable of expressing long-term potentiation (LTP). The C-terminal domain (CT) of GluD1 has a classic PDZ-binding motif, which is critical for the synaptic trafficking of other glutamate receptors, but we found that its binding to PSD-95 was very weak, and deleting the PDZ-binding motif failed to alter synaptic trafficking. However, deletion of the entire CT abolished synaptic trafficking, but not surface expression. We found that mutation of threonine (T) T923 to an alanine disrupted synaptic trafficking. Therefore, GluD1 receptors have strikingly different trafficking mechanisms compared with AMPARs. These results highlight the diversity of ionotropic glutamate receptor trafficking rules at a single type of synapse. Since this receptor is genetically associated with schizophrenia, our findings may provide an important clue to understand schizophrenia.
- Published
- 2019
26. The Golgi Outpost Protein TPPP Nucleates Microtubules and Is Critical for Myelination
- Author
-
Fu, Meng-Meng, McAlear, Thomas S, Nguyen, Huy, Oses-Prieto, Juan A, Valenzuela, Alex, Shi, Rebecca D, Perrino, John J, Huang, Ting-Ting, Burlingame, Alma L, Bechstedt, Susanne, and Barres, Ben A
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Brain Disorders ,Neurosciences ,Underpinning research ,1.1 Normal biological development and functioning ,Animals ,Animals ,Newborn ,Axons ,Carrier Proteins ,Cell-Free System ,Cells ,Cultured ,Escherichia coli ,Gene Knockdown Techniques ,Golgi Apparatus ,Humans ,Mice ,Mice ,Inbred C57BL ,Mice ,Knockout ,Microtubule-Organizing Center ,Microtubules ,Myelin Sheath ,Nerve Tissue Proteins ,Oligodendrocyte Precursor Cells ,Rats ,Rats ,Sprague-Dawley ,Tubulin ,Golgi outpost ,TPPP ,branching ,mass spectrometry ,microtubule ,myelin ,nucleation ,oligodendrocyte ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
Oligodendrocytes extend elaborate microtubule arbors that contact up to 50 axon segments per cell, then spiral around myelin sheaths, penetrating from outer to inner layers. However, how they establish this complex cytoarchitecture is unclear. Here, we show that oligodendrocytes contain Golgi outposts, an organelle that can function as an acentrosomal microtubule-organizing center (MTOC). We identify a specific marker for Golgi outposts-TPPP (tubulin polymerization promoting protein)-that we use to purify this organelle and characterize its proteome. In in vitro cell-free assays, recombinant TPPP nucleates microtubules. Primary oligodendrocytes from Tppp knockout (KO) mice have aberrant microtubule branching, mixed microtubule polarity, and shorter myelin sheaths when cultured on 3-dimensional (3D) microfibers. Tppp KO mice exhibit hypomyelination with shorter, thinner myelin sheaths and motor coordination deficits. Together, our data demonstrate that microtubule nucleation outside the cell body at Golgi outposts by TPPP is critical for elongation of the myelin sheath.
- Published
- 2019
27. The Gene-Silencing Protein MORC-1 Topologically Entraps DNA and Forms Multimeric Assemblies to Cause DNA Compaction
- Author
-
Kim, HyeongJun, Yen, Linda, Wongpalee, Somsakul P, Kirshner, Jessica A, Mehta, Nicita, Xue, Yan, Johnston, Jonathan B, Burlingame, Alma L, Kim, John K, Loparo, Joseph J, and Jacobsen, Steve E
- Subjects
Biochemistry and Cell Biology ,Bioinformatics and Computational Biology ,Biological Sciences ,Genetics ,Underpinning research ,1.1 Normal biological development and functioning ,Generic health relevance ,Animals ,Caenorhabditis elegans ,Caenorhabditis elegans Proteins ,DNA ,Helminth ,Nuclear Proteins ,Nucleic Acid Conformation ,Nucleosomes ,Protein Multimerization ,DNA compaction ,DNA-binding protein ,GHKL ATPases ,MORC ,genome organization ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
Microrchidia (MORC) ATPases are critical for gene silencing and chromatin compaction in multiple eukaryotic systems, but the mechanisms by which MORC proteins act are poorly understood. Here, we apply a series of biochemical, single-molecule, and cell-based imaging approaches to better understand the function of the Caenorhabditis elegans MORC-1 protein. We find that MORC-1 binds to DNA in a length-dependent but sequence non-specific manner and compacts DNA by forming DNA loops. MORC-1 molecules diffuse along DNA but become static as they grow into foci that are topologically entrapped on DNA. Consistent with the observed MORC-1 multimeric assemblies, MORC-1 forms nuclear puncta in cells and can also form phase-separated droplets in vitro. We also demonstrate that MORC-1 compacts nucleosome templates. These results suggest that MORCs affect genome structure and gene silencing by forming multimeric assemblages to topologically entrap and progressively loop and compact chromatin.
- Published
- 2019
28. Tau/MAPT disease-associated variant A152T alters tau function and toxicity via impaired retrograde axonal transport.
- Author
-
Butler, Victoria J, Salazar, Dominique A, Soriano-Castell, David, Alves-Ferreira, Miguel, Dennissen, Frank JA, Vohra, Mihir, Oses-Prieto, Juan A, Li, Kathy H, Wang, Austin L, Jing, Beibei, Li, Biao, Groisman, Alex, Gutierrez, Edgar, Mooney, Sean, Burlingame, Alma L, Ashrafi, Kaveh, Mandelkow, Eva-Maria, Encalada, Sandra E, and Kao, Aimee W
- Subjects
Axons ,Synaptic Vesicles ,Animals ,Animals ,Genetically Modified ,Humans ,Mice ,Caenorhabditis elegans ,Tauopathies ,Disease Models ,Animal ,Disease Susceptibility ,tau Proteins ,Amino Acid Substitution ,Axonal Transport ,Protein Binding ,Phosphorylation ,Mutation ,Alleles ,Genetic Variation ,Genetically Modified ,Disease Models ,Animal ,Genetics & Heredity ,Biological Sciences ,Medical and Health Sciences - Abstract
Mutations in the microtubule-associated protein tau (MAPT) underlie multiple neurodegenerative disorders, yet the pathophysiological mechanisms are unclear. A novel variant in MAPT resulting in an alanine to threonine substitution at position 152 (A152T tau) has recently been described as a significant risk factor for both frontotemporal lobar degeneration and Alzheimer's disease. Here we use complementary computational, biochemical, molecular, genetic and imaging approaches in Caenorhabditis elegans and mouse models to interrogate the effects of the A152T variant on tau function. In silico analysis suggests that a threonine at position 152 of tau confers a new phosphorylation site. This finding is borne out by mass spectrometric survey of A152T tau phosphorylation in C. elegans and mouse. Optical pulse-chase experiments of Dendra2-tau demonstrate that A152T tau and phosphomimetic A152E tau exhibit increased diffusion kinetics and the ability to traverse across the axon initial segment more efficiently than wild-type (WT) tau. A C. elegans model of tauopathy reveals that A152T and A152E tau confer patterns of developmental toxicity distinct from WT tau, likely due to differential effects on retrograde axonal transport. These data support a role for phosphorylation of the variant threonine in A152T tau toxicity and suggest a mechanism involving impaired retrograde axonal transport contributing to human neurodegenerative disease.
- Published
- 2019
29. Phosphoregulation of the oncogenic protein regulator of cytokinesis 1 (PRC1) by the atypical CDK16/CCNY complex.
- Author
-
Hernández-Ortega, Sara, Sánchez-Botet, Abril, Quandt, Eva, Masip, Núria, Gasa, Laura, Verde, Gaetano, Jiménez, Javier, Levin, Rebecca S, Rutaganira, Florentine U, Burlingame, Alma L, Wolfgeher, Don, Ribeiro, Mariana PC, Kron, Stephen J, Shokat, Kevan M, and Clotet, Josep
- Subjects
Cell Line ,Hela Cells ,Humans ,Cyclin-Dependent Kinases ,Cell Cycle Proteins ,Cell Division ,Protein Binding ,Phosphorylation ,HEK293 Cells ,MCF-7 Cells ,Clustered Regularly Interspaced Short Palindromic Repeats ,HeLa Cells ,Biotechnology ,Cancer ,2.1 Biological and endogenous factors ,Medicinal and Biomolecular Chemistry ,Biochemistry & Molecular Biology - Abstract
CDK16 (also known as PCTAIRE1 or PCTK1) is an atypical member of the cyclin-dependent kinase (CDK) family that forms an active complex with cyclin Y (CCNY). Although both proteins have been recently implicated in cancer pathogenesis, it is still unclear how the CDK16/CCNY complex exerts its biological activity. To understand the CDK16/CCNY network, we used complementary proteomic approaches to identify potential substrates of this complex. We identified several candidates implicating the CDK16/CCNY complex in cytoskeletal dynamics, and we focused on the microtubule-associated protein regulator of cytokinesis (PRC1), an essential protein for cell division that organizes antiparallel microtubules and whose deregulation may drive genomic instability in cancer. Using analog-sensitive (AS) CDK16 generated by CRISPR-Cas9 mutagenesis in 293T cells, we found that specific inhibition of CDK16 induces PRC1 dephosphorylation at Thr481 and delocalization to the nucleus during interphase. The observation that CDK16 inhibition and PRC1 downregulation exhibit epistatic effects on cell viability confirms that these proteins can act through a single pathway. In conclusion, we identified PRC1 as the first substrate of the CDK16/CCNY complex and demonstrated that the proliferative function of CDK16 is mediated by PRC1 phosphorylation. As CDK16 is emerging as a critical node in cancer, our study reveals novel potential therapeutic targets.
- Published
- 2019
30. Chemically induced vesiculation as a platform for studying TMEM16F activity
- Author
-
Han, Tina W, Ye, Wenlei, Bethel, Neville P, Zubia, Mario, Kim, Andrew, Li, Kathy H, Burlingame, Alma L, Grabe, Michael, Jan, Yuh Nung, and Jan, Lily Y
- Subjects
Biochemistry and Cell Biology ,Medical Physiology ,Biomedical and Clinical Sciences ,Biological Sciences ,Prevention ,Underpinning research ,1.1 Normal biological development and functioning ,Generic health relevance ,Animals ,Anoctamins ,Biological Transport ,Calcium ,Cell Line ,Cell Line ,Tumor ,Cell Membrane ,Cell-Derived Microparticles ,Extracellular Vesicles ,HEK293 Cells ,Humans ,Mice ,Phospholipid Transfer Proteins ,Phospholipids ,TMEM16F ,phospholipid scrambling ,extracellular vesicles ,calcium influx ,GPMV - Abstract
Calcium-activated phospholipid scramblase mediates the energy-independent bidirectional translocation of lipids across the bilayer, leading to transient or, in the case of apoptotic scrambling, sustained collapse of membrane asymmetry. Cells lacking TMEM16F-dependent lipid scrambling activity are deficient in generation of extracellular vesicles (EVs) that shed from the plasma membrane in a Ca2+-dependent manner, namely microvesicles. We have adapted chemical induction of giant plasma membrane vesicles (GPMVs), which require both TMEM16F-dependent phospholipid scrambling and calcium influx, as a kinetic assay to investigate the mechanism of TMEM16F activity. Using the GPMV assay, we identify and characterize both inactivating and activating mutants that elucidate the mechanism for TMEM16F activation and facilitate further investigation of TMEM16F-mediated lipid translocation and its role in extracellular vesiculation.
- Published
- 2019
31. The lineage stability and suppressive program of regulatory T cells require protein O-GlcNAcylation.
- Author
-
Liu, Bing, Salgado, Oscar C, Singh, Sangya, Hippen, Keli L, Maynard, Jason C, Burlingame, Alma L, Ball, Lauren E, Blazar, Bruce R, Farrar, Michael A, Hogquist, Kristin A, and Ruan, Hai-Bin
- Subjects
Animals ,Mice ,Transgenic ,Humans ,Mice ,Acetylglucosamine ,Receptors ,Antigen ,T-Cell ,Interleukin-2 ,Signal Transduction ,Autoimmunity ,Self Tolerance ,Protein Processing ,Post-Translational ,Cell Lineage ,Genes ,Reporter ,Female ,Male ,T-Lymphocytes ,Regulatory ,STAT5 Transcription Factor ,Forkhead Transcription Factors ,Primary Cell Culture ,Emerging Infectious Diseases ,Prevention ,Biodefense ,Autoimmune Disease ,Vaccine Related ,2.1 Biological and endogenous factors ,1.1 Normal biological development and functioning ,Inflammatory and Immune System ,Transgenic ,Receptors ,Antigen ,T-Cell ,Protein Processing ,Post-Translational ,Genes ,Reporter ,T-Lymphocytes ,Regulatory ,MD Multidisciplinary - Abstract
Regulatory T (Treg) cells control self-tolerance, inflammatory responses and tissue homeostasis. In mature Treg cells, continued expression of FOXP3 maintains lineage identity, while T cell receptor (TCR) signaling and interleukin-2 (IL-2)/STAT5 activation support the suppressive effector function of Treg cells, but how these regulators synergize to control Treg cell homeostasis and function remains unclear. Here we show that TCR-activated posttranslational modification by O-linked N-Acetylglucosamine (O-GlcNAc) stabilizes FOXP3 and activates STAT5, thus integrating these critical signaling pathways. O-GlcNAc-deficient Treg cells develop normally but display modestly reduced FOXP3 expression, strongly impaired lineage stability and effector function, and ultimately fatal autoimmunity in mice. Moreover, deficiency in protein O-GlcNAcylation attenuates IL-2/STAT5 signaling, while overexpression of a constitutively active form of STAT5 partially ameliorates Treg cell dysfunction and systemic inflammation in O-GlcNAc deficient mice. Collectively, our data demonstrate that protein O-GlcNAcylation is essential for lineage stability and effector function in Treg cells.
- Published
- 2019
32. Histone H3 binding to the PHD1 domain of histone demethylase KDM5A enables active site remodeling.
- Author
-
Longbotham, James E, Chio, Cynthia M, Dharmarajan, Venkatasubramanian, Trnka, Michael J, Torres, Idelisse Ortiz, Goswami, Devrishi, Ruiz, Karen, Burlingame, Alma L, Griffin, Patrick R, and Fujimori, Danica Galonić
- Subjects
Animals ,Histones ,Catalytic Domain ,Protein Conformation ,Models ,Molecular ,Retinoblastoma-Binding Protein 2 ,Sf9 Cells ,Protein Domains ,Cancer ,Genetics ,2.1 Biological and endogenous factors ,Models ,Molecular ,MD Multidisciplinary - Abstract
Histone demethylase KDM5A removes methyl marks from lysine 4 of histone H3 and is often overexpressed in cancer. The in vitro demethylase activity of KDM5A is allosterically enhanced by binding of its product, unmodified H3 peptides, to its PHD1 reader domain. However, the molecular basis of this allosteric enhancement is unclear. Here we show that saturation of the PHD1 domain by the H3 N-terminal tail peptides stabilizes binding of the substrate to the catalytic domain and improves the catalytic efficiency of demethylation. When present in saturating concentrations, differently modified H3 N-terminal tail peptides have a similar effect on demethylation. However, they vary greatly in their affinity towards the PHD1 domain, suggesting that H3 modifications can tune KDM5A activity. Furthermore, hydrogen/deuterium exchange coupled with mass spectrometry (HDX-MS) experiments reveal conformational changes in the allosterically enhanced state. Our findings may enable future development of anti-cancer therapies targeting regions involved in allosteric regulation.
- Published
- 2019
33. A New Monoclonal Antibody Enables BAR Analysis of Subcellular Importin β1 Interactomes
- Author
-
Song, Didi-Andreas, Alber, Stefanie, Doron-Mandel, Ella, Schmid, Vera, Albus, Christin A., Leitner, Orith, Hamawi, Hedva, Oses-Prieto, Juan A., Dezorella, Nili, Burlingame, Alma L., Fainzilber, Mike, and Rishal, Ida
- Published
- 2022
- Full Text
- View/download PDF
34. Deconvoluting signals downstream of growth and immune receptor kinases by phosphocodes of the BSU1 family phosphatases
- Author
-
Park, Chan Ho, Bi, Yang, Youn, Ji-Hyun, Kim, So-Hee, Kim, Jung-Gun, Xu, Nicole Y., Shrestha, Ruben, Burlingame, Alma L., Xu, Shou-Ling, Mudgett, Mary Beth, Kim, Seong-Ki, Kim, Tae-Wuk, and Wang, Zhi-Yong
- Published
- 2022
- Full Text
- View/download PDF
35. Epithelial STAT6 O-GlcNAcylation drives a concerted anti-helminth alarmin response dependent on tuft cell hyperplasia and Gasdermin C
- Author
-
Zhao, Ming, Ren, Kaiqun, Xiong, Xiwen, Xin, Yue, Zou, Yujie, Maynard, Jason C., Kim, Angela, Battist, Alexander P., Koneripalli, Navya, Wang, Yusu, Chen, Qianyue, Xin, Ruyue, Yang, Chenyan, Huang, Rong, Yu, Jiahui, Huang, Zan, Zhang, Zengdi, Wang, Haiguang, Wang, Daoyuan, Xiao, Yihui, Salgado, Oscar C., Jarjour, Nicholas N., Hogquist, Kristin A., Revelo, Xavier S., Burlingame, Alma L., Gao, Xiang, von Moltke, Jakob, Lin, Zhaoyu, and Ruan, Hai-Bin
- Published
- 2022
- Full Text
- View/download PDF
36. TRIM46 Is Required for Microtubule Fasciculation In Vivo But Not Axon Specification or Axon Initial Segment Formation.
- Author
-
Melton, Allison J., Palfini, Victoria L., Yuki Ogawa, Oses Prieto, Juan A., Vainshtein, Anna, Burlingame, Alma L., Peles, Elior, and Rasband, Matthew N.
- Subjects
TRIM proteins ,KNOCKOUT mice ,ACTION potentials ,NERVOUS system ,MICROTUBULE-associated proteins - Abstract
Vertebrate nervous systems use the axon initial segment (AIS) to initiate action potentials and maintain neuronal polarity. The microtubule-associated protein tripartite motif containing 46 (TRIM46) was reported to regulate axon specification, AIS assembly, and neuronal polarity through the bundling, or fasciculation, of microtubules in the proximal axon. However, these claims are based on TRIM46 knockdown in cultured neurons. To investigate TRIM46 function in vivo, we examined male and female TRIM46 knock-out mice. Contrary to previous reports, we find that TRIM46 is dispensable for axon specification and AIS formation. TRIM46 knock-out mice are viable, have normal behavior, and have normal brain structure. Thus, TRIM46 is not required for AIS formation, axon specification, or nervous system function. However, we confirm that TRIM46 is required for microtubule fasciculation. We also show TRIM46 enrichment in the first ~100 µm of axon occurs independently of ankyrinG (AnkG) in vivo, although AnkG is required to restrict TRIM46 only to the AIS. Our results highlight the need for further investigation of the mechanisms by which the AIS and microtubules interact to shape neuronal structure and function. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
37. hnRNPs Interacting with mRNA Localization Motifs Define Axonal RNA Regulons.
- Author
-
Lee, Seung Joon, Oses-Prieto, Juan A, Kawaguchi, Riki, Sahoo, Pabitra K, Kar, Amar N, Rozenbaum, Meir, Oliver, David, Chand, Shreya, Ji, Hao, Shtutman, Michael, Miller-Randolph, Sharmina, Taylor, Ross J, Fainzilber, Mike, Coppola, Giovanni, Burlingame, Alma L, and Twiss, Jeffery L
- Subjects
Axons ,Animals ,Rats ,Sprague-Dawley ,Neuropeptides ,Heterogeneous-Nuclear Ribonucleoproteins ,HMGB1 Protein ,GAP-43 Protein ,RNA ,Messenger ,5' Untranslated Regions ,Axonal Transport ,Protein Biosynthesis ,Protein Binding ,RNA Transport ,Regulon ,Male ,GPI-Linked Proteins ,Nucleotide Motifs ,Affinity proteomics ,Bioinformatics ,Neurobiology ,RNA binding protein ,Ribonucleoproteins ,Subcellular analysis ,axon growth ,hnRNP ,Genetics ,Neurosciences ,1.1 Normal biological development and functioning ,Underpinning research ,Neurological ,Biochemistry & Molecular Biology - Abstract
mRNA translation in axons enables neurons to introduce new proteins at sites distant from their cell body. mRNA-protein interactions drive this post-transcriptional regulation, yet knowledge of RNA binding proteins (RBP) in axons is limited. Here we used proteomics to identify RBPs interacting with the axonal localizing motifs of Nrn1, Hmgb1, Actb, and Gap43 mRNAs, revealing many novel RBPs in axons. Interestingly, no RBP is shared between all four RNA motifs, suggesting graded and overlapping specificities of RBP-mRNA pairings. A systematic assessment of axonal mRNAs interacting with hnRNP H1, hnRNP F, and hnRNP K, proteins that bound with high specificity to Nrn1 and Hmgb1, revealed that axonal mRNAs segregate into axon growth-associated RNA regulons based on hnRNP interactions. Axotomy increases axonal transport of hnRNPs H1, F, and K, depletion of these hnRNPs decreases axon growth and reduces axonal mRNA levels and axonal protein synthesis. Thus, subcellular hnRNP-interacting RNA regulons support neuronal growth and regeneration.
- Published
- 2018
38. A Viral Protein Restricts Drosophila RNAi Immunity by Regulating Argonaute Activity and Stability
- Author
-
Nayak, Arabinda, Kim, Dong Young, Trnka, Michael J, Kerr, Craig H, Lidsky, Peter V, Stanley, David J, Rivera, Brianna Monique, Li, Kathy H, Burlingame, Alma L, Jan, Eric, Frydman, Judith, Gross, John D, and Andino, Raul
- Subjects
Emerging Infectious Diseases ,Genetics ,Infectious Diseases ,Biotechnology ,Aetiology ,2.2 Factors relating to the physical environment ,Infection ,Animals ,Argonaute Proteins ,Cell Line ,Dicistroviridae ,Drosophila Proteins ,Drosophila melanogaster ,Humans ,Mutation ,Protein Binding ,Protein Conformation ,Protein Interaction Maps ,RNA Interference ,Ubiquitin-Protein Ligases ,Viral Proteins ,Virus Replication ,Ago-2 ,Ago-2 degradation ,CrPV ,E3 ligase ,RNAi ,RNAi suppressor ,antiviral immunity ,insects ,Microbiology ,Medical Microbiology ,Immunology - Abstract
The dicistrovirus, Cricket paralysis virus (CrPV) encodes an RNA interference (RNAi) suppressor, 1A, which modulates viral virulence. Using the Drosophila model, we combined structural, biochemical, and virological approaches to elucidate the strategies by which CrPV-1A restricts RNAi immunity. The atomic resolution structure of CrPV-1A uncovered a flexible loop that interacts with Argonaute 2 (Ago-2), thereby inhibiting Ago-2 endonuclease-dependent immunity. Mutations disrupting Ago-2 binding attenuates viral pathogenesis in wild-type but not Ago-2-deficient flies. CrPV-1A also contains a BC-box motif that enables the virus to hijack a host Cul2-Rbx1-EloBC ubiquitin ligase complex, which promotes Ago-2 degradation and virus replication. Our study uncovers a viral-based dual regulatory program that restricts antiviral immunity by direct interaction with and modulation of host proteins. While the direct inhibition of Ago-2 activity provides an efficient mechanism to establish infection, the recruitment of a ubiquitin ligase complex enables CrPV-1A to amplify Ago-2 inactivation to restrict further antiviral RNAi immunity.
- Published
- 2018
39. Targeting CD46 for both adenocarcinoma and neuroendocrine prostate cancer.
- Author
-
Su, Yang, Liu, Yue, Behrens, Christopher R, Bidlingmaier, Scott, Lee, Nam-Kyung, Aggarwal, Rahul, Sherbenou, Daniel W, Burlingame, Alma L, Hann, Byron C, Simko, Jeffry P, Premasekharan, Gayatri, Paris, Pamela L, Shuman, Marc A, Seo, Youngho, Small, Eric J, and Liu, Bin
- Subjects
Prostate ,Cell Line ,Tumor ,Animals ,Macaca fascicularis ,Humans ,Neuroendocrine Tumors ,Adenocarcinoma ,Prostatic Neoplasms ,Benzamides ,Nitriles ,Phenylthiohydantoin ,Androstenes ,Recombinant Fusion Proteins ,Antineoplastic Agents ,Antibodies ,Monoclonal ,Antibodies ,Neoplasm ,Antigens ,Neoplasm ,Therapeutics ,Xenograft Model Antitumor Assays ,Signal Transduction ,Antibody Affinity ,Female ,Male ,Tumor Microenvironment ,Membrane Cofactor Protein ,Oncology ,Prostate cancer ,Cancer ,Rare Diseases ,Prostate Cancer ,Biotechnology ,Urologic Diseases ,5.1 Pharmaceuticals ,Development of treatments and therapeutic interventions - Abstract
Although initially responsive to androgen signaling inhibitors (ASIs), metastatic castration-resistant prostate cancer (mCRPC) inevitably develops and is incurable. In addition to adenocarcinoma (adeno), neuroendocrine prostate cancer (NEPC) emerges to confer ASI resistance. We have previously combined laser capture microdissection and phage antibody display library selection on human cancer specimens and identified novel internalizing antibodies binding to tumor cells residing in their tissue microenvironment. We identified the target antigen for one of these antibodies as CD46, a multifunctional protein that is best known for negatively regulating the innate immune system. CD46 is overexpressed in primary tumor tissue and CRPC (localized and metastatic; adeno and NEPC), but expressed at low levels on normal tissues except for placental trophoblasts and prostate epithelium. Abiraterone- and enzalutamide-treated mCRPC cells upregulate cell surface CD46 expression. Genomic analysis showed that the CD46 gene is gained in 45% abiraterone-resistant mCRPC patients. We conjugated a tubulin inhibitor to our macropinocytosing anti-CD46 antibody and showed that the resulting antibody-drug conjugate (ADC) potently and selectively kills both adeno and NEPC cell lines in vitro (sub-nM EC50) but not normal cells. CD46 ADC regressed and eliminated an mCRPC cell line xenograft in vivo in both subcutaneous and intrafemoral models. Exploratory toxicology studies of the CD46 ADC in non-human primates demonstrated an acceptable safety profile. Thus, CD46 is an excellent target for antibody-based therapy development, which has potential to be applicable to both adenocarcinoma and neuroendocrine types of mCRPC that are resistant to current treatment.
- Published
- 2018
40. Axonal G3BP1 stress granule protein limits axonal mRNA translation and nerve regeneration.
- Author
-
Sahoo, Pabitra K, Lee, Seung Joon, Jaiswal, Poonam B, Alber, Stefanie, Kar, Amar N, Miller-Randolph, Sharmina, Taylor, Elizabeth E, Smith, Terika, Singh, Bhagat, Ho, Tammy Szu-Yu, Urisman, Anatoly, Chand, Shreya, Pena, Edsel A, Burlingame, Alma L, Woolf, Clifford J, Fainzilber, Mike, English, Arthur W, and Twiss, Jeffery L
- Subjects
Axons ,Cells ,Cultured ,NIH 3T3 Cells ,Cytoplasmic Granules ,Animals ,Humans ,Mice ,Rats ,Rats ,Sprague-Dawley ,RNA ,Messenger ,Microscopy ,Fluorescence ,Fluorescence Recovery After Photobleaching ,Nerve Regeneration ,Female ,Male ,HEK293 Cells ,Poly-ADP-Ribose Binding Proteins - Abstract
Critical functions of intra-axonally synthesized proteins are thought to depend on regulated recruitment of mRNA from storage depots in axons. Here we show that axotomy of mammalian neurons induces translation of stored axonal mRNAs via regulation of the stress granule protein G3BP1, to support regeneration of peripheral nerves. G3BP1 aggregates within peripheral nerve axons in stress granule-like structures that decrease during regeneration, with a commensurate increase in phosphorylated G3BP1. Colocalization of G3BP1 with axonal mRNAs is also correlated with the growth state of the neuron. Disrupting G3BP functions by overexpressing a dominant-negative protein activates intra-axonal mRNA translation, increases axon growth in cultured neurons, disassembles axonal stress granule-like structures, and accelerates rat nerve regeneration in vivo.
- Published
- 2018
41. Schwann cell O-GlcNAcylation promotes peripheral nerve remyelination via attenuation of the AP-1 transcription factor JUN
- Author
-
Kim, Sungsu, Maynard, Jason C, Strickland, Amy, Burlingame, Alma L, and Milbrandt, Jeffrey
- Subjects
Neurosciences ,Regenerative Medicine ,Autoimmune Disease ,Genetics ,Neurodegenerative ,Peripheral Neuropathy ,Physical Injury - Accidents and Adverse Effects ,1.1 Normal biological development and functioning ,Underpinning research ,Aetiology ,2.1 Biological and endogenous factors ,Neurological ,Acylation ,Aging ,Animals ,Axons ,Demyelinating Diseases ,Diabetic Neuropathies ,Gene Deletion ,Mice ,Mice ,Knockout ,N-Acetylglucosaminyltransferases ,Nerve Regeneration ,Oncogene Protein p65(gag-jun) ,Schwann Cells ,Sciatic Nerve ,Transcription Factor AP-1 ,OGT ,Schwann cells ,O-GlcNAcylation ,JUN ,nerve injury - Abstract
Schwann cells (SCs), the glia of the peripheral nervous system, play an essential role in nerve regeneration. Upon nerve injury, SCs are reprogrammed into unique "repair SCs," and these cells remove degenerating axons/myelin debris, promote axonal regrowth, and ultimately remyelinate regenerating axons. The AP-1 transcription factor JUN is promptly induced in SCs upon nerve injury and potently mediates this injury-induced SC plasticity; however, the regulation of these JUN-dependent SC injury responses is unclear. Previously, we produced mice with a SC-specific deletion of O-GlcNAc transferase (OGT). This enzyme catalyzes O-GlcNAcylation, a posttranslational modification that is influenced by the cellular metabolic state. Mice lacking OGT in SCs develop a progressive demyelinating peripheral neuropathy. Here, we investigated the nerve repair process in OGT-SCKO mutant mice and found that the remyelination of regenerating axons is severely impaired. Gene expression profiling of OGT-SCKO SCs revealed that the JUN-dependent SC injury program was elevated in the absence of injury and failed to shut down at the appropriate time after injury. This aberrant JUN activity results in abnormalities in repair SC function and redifferentiation and prevents the timely remyelination. This aberrant nerve injury response is normalized in OGT-SCKO mice with reduced Jun gene dosage in SCs. Mechanistically, OGT O-GlcNAcylates JUN at multiple sites, which then leads to an attenuation of AP-1 transcriptional activity. Together, these results highlight the metabolic oversight of the nerve injury response via the regulation of JUN activity by O-GlcNAcylation, a pathway that could be important in the neuropathy associated with diabetes and aging.
- Published
- 2018
42. Prp8 positioning of U5 snRNA is linked to 5′ splice site recognition
- Author
-
MacRae, Andrew J, Mayerle, Megan, Hrabeta-Robinson, Eva, Chalkley, Robert J, Guthrie, Christine, Burlingame, Alma L, and Jurica, Melissa S
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Genetics ,Neurosciences ,Generic health relevance ,Catalytic Domain ,Humans ,RNA Precursors ,RNA Splice Sites ,RNA ,Small Nuclear ,RNA-Binding Proteins ,Ribonucleoprotein ,U4-U6 Small Nuclear ,Ribonucleoprotein ,U5 Small Nuclear ,Saccharomyces cerevisiae ,Spliceosomes ,pre-mRNA splicing ,spliceosome ,Prp8 ,U5 snRNA ,' exon ,chemical probing ,5′ exon ,Developmental Biology ,Biochemistry and cell biology - Abstract
Prp8 is an essential protein that regulates spliceosome assembly and conformation during pre-mRNA splicing. Recent cryo-EM structures of the spliceosome model Prp8 as a scaffold for the spliceosome's catalytic U snRNA components. Using a new amino acid probing strategy, we identified a dynamic region in human Prp8 that is positioned to stabilize the pre-mRNA in the spliceosome active site through interactions with U5 snRNA. Mutagenesis of the identified Prp8 residues in yeast indicates a role in 5' splice site recognition. Genetic interactions with spliceosome proteins Isy1, which buttresses the intron branch point, and Snu114, a regulatory GTPase that directly contacts Prp8, further corroborate a role for the same Prp8 residues in substrate positioning and activation. Together the data suggest that adjustments in interactions between Prp8 and U5 snRNA help establish proper positioning of the pre-mRNA into the active site to enhance 5' splice site fidelity.
- Published
- 2018
43. The ER membrane protein complex interacts cotranslationally to enable biogenesis of multipass membrane proteins.
- Author
-
Shurtleff, Matthew J, Itzhak, Daniel N, Hussmann, Jeffrey A, Schirle Oakdale, Nicole T, Costa, Elizabeth A, Jonikas, Martin, Weibezahn, Jimena, Popova, Katerina D, Jan, Calvin H, Sinitcyn, Pavel, Vembar, Shruthi S, Hernandez, Hilda, Cox, Jürgen, Burlingame, Alma L, Brodsky, Jeffrey L, Frost, Adam, Borner, Georg Hh, and Weissman, Jonathan S
- Subjects
Endoplasmic Reticulum ,Ribosomes ,Humans ,Saccharomyces cerevisiae ,Multiprotein Complexes ,Membrane Proteins ,Molecular Chaperones ,Proteomics ,Protein Biosynthesis ,Protein Transport ,EMC ,cell biology ,endoplasmic reticulum ,human ,ion channel ,transmembrane ,transporter ,Biochemistry and Cell Biology - Abstract
The endoplasmic reticulum (ER) supports biosynthesis of proteins with diverse transmembrane domain (TMD) lengths and hydrophobicity. Features in transmembrane domains such as charged residues in ion channels are often functionally important, but could pose a challenge during cotranslational membrane insertion and folding. Our systematic proteomic approaches in both yeast and human cells revealed that the ER membrane protein complex (EMC) binds to and promotes the biogenesis of a range of multipass transmembrane proteins, with a particular enrichment for transporters. Proximity-specific ribosome profiling demonstrates that the EMC engages clients cotranslationally and immediately following clusters of TMDs enriched for charged residues. The EMC can remain associated after completion of translation, which both protects clients from premature degradation and allows recruitment of substrate-specific and general chaperones. Thus, the EMC broadly enables the biogenesis of multipass transmembrane proteins containing destabilizing features, thereby mitigating the trade-off between function and stability.
- Published
- 2018
44. A molecular cascade modulates MAP1B and confers resistance to mTOR inhibition in human glioblastoma
- Author
-
Laks, Dan R, Oses-Prieto, Juan A, Alvarado, Alvaro G, Nakashima, Jonathan, Chand, Shreya, Azzam, Daniel B, Gholkar, Ankur A, Sperry, Jantzen, Ludwig, Kirsten, Condro, Michael C, Nazarian, Serli, Cardenas, Anjelica, Shih, Michelle YS, Damoiseaux, Robert, France, Bryan, Orozco, Nicholas, Visnyei, Koppany, Crisman, Thomas J, Gao, Fuying, Torres, Jorge Z, Coppola, Giovanni, Burlingame, Alma L, and Kornblum, Harley I
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Rare Diseases ,Brain Disorders ,Cancer ,Neurosciences ,Emerging Infectious Diseases ,Brain Cancer ,5.1 Pharmaceuticals ,Development of treatments and therapeutic interventions ,Animals ,Antibiotics ,Antineoplastic ,Apoptosis ,Biomarkers ,Tumor ,Cell Proliferation ,Extracellular Signal-Regulated MAP Kinases ,Gene Expression Regulation ,Neoplastic ,Glioblastoma ,Glycogen Synthase Kinase 3 beta ,Humans ,Male ,Mice ,Mice ,Inbred NOD ,Mice ,SCID ,Microtubule-Associated Proteins ,Phosphatidylinositol 3-Kinases ,Phosphoinositide-3 Kinase Inhibitors ,Phosphorylation ,Proto-Oncogene Proteins c-akt ,RNA ,Small Interfering ,Signal Transduction ,Sirolimus ,TOR Serine-Threonine Kinases ,Tumor Cells ,Cultured ,Xenograft Model Antitumor Assays ,cancer ,ERK ,glioma ,glioblastoma ,GSK3B ,MAP1B ,MEK ,microtubule ,mTOR ,tubulin ,Oncology & Carcinogenesis ,Oncology and carcinogenesis - Abstract
BackgroundClinical trials of therapies directed against nodes of the signaling axis of phosphatidylinositol-3 kinase/Akt/mammalian target of rapamycin (mTOR) in glioblastoma (GBM) have had disappointing results. Resistance to mTOR inhibitors limits their efficacy.MethodsTo determine mechanisms of resistance to chronic mTOR inhibition, we performed tandem screens on patient-derived GBM cultures.ResultsAn unbiased phosphoproteomic screen quantified phosphorylation changes associated with chronic exposure to the mTOR inhibitor rapamycin, and our analysis implicated a role for glycogen synthase kinase (GSK)3B attenuation in mediating resistance that was confirmed by functional studies. A targeted short hairpin RNA screen and further functional studies both in vitro and in vivo demonstrated that microtubule-associated protein (MAP)1B, previously associated predominantly with neurons, is a downstream effector of GSK3B-mediated resistance. Furthermore, we provide evidence that chronic rapamycin induces microtubule stability in a MAP1B-dependent manner in GBM cells. Additional experiments explicate a signaling pathway wherein combinatorial extracellular signal-regulated kinase (ERK)/mTOR targeting abrogates inhibitory phosphorylation of GSK3B, leads to phosphorylation of MAP1B, and confers sensitization.ConclusionsThese data portray a compensatory molecular signaling network that imparts resistance to chronic mTOR inhibition in primary, human GBM cell cultures and points toward new therapeutic strategies.
- Published
- 2018
45. The nucleosomal acidic patch relieves auto-inhibition by the ISWI remodeler SNF2h.
- Author
-
Gamarra, Nathan, Johnson, Stephanie L, Trnka, Michael J, Burlingame, Alma L, and Narlikar, Geeta J
- Subjects
Nucleosomes ,Humans ,Chromosomal Proteins ,Non-Histone ,Histones ,Fluorescence Resonance Energy Transfer ,Adenosine Triphosphatases ,Single Molecule Imaging ,ATP-Dependent Chromatin Remodeling ,INO80 ,ISWI ,S. cerevisiae ,chromatin ,chromosomes ,cross-linking mass spectrometry ,gene expression ,human ,molecular biophysics ,smFRET ,structural biology ,Chromosomal Proteins ,Non-Histone ,1.1 Normal biological development and functioning ,Biochemistry and Cell Biology - Abstract
ISWI family chromatin remodeling motors use sophisticated autoinhibition mechanisms to control nucleosome sliding. Yet how the different autoinhibitory domains are regulated is not well understood. Here we show that an acidic patch formed by histones H2A and H2B of the nucleosome relieves the autoinhibition imposed by the AutoN and the NegC regions of the human ISWI remodeler SNF2h. Further, by single molecule FRET we show that the acidic patch helps control the distance travelled per translocation event. We propose a model in which the acidic patch activates SNF2h by providing a landing pad for the NegC and AutoN auto-inhibitory domains. Interestingly, the INO80 complex is also strongly dependent on the acidic patch for nucleosome sliding, indicating that this substrate feature can regulate remodeling enzymes with substantially different mechanisms. We therefore hypothesize that regulating access to the acidic patch of the nucleosome plays a key role in coordinating the activities of different remodelers in the cell.
- Published
- 2018
46. Locally translated mTOR controls axonal local translation in nerve injury
- Author
-
Terenzio, Marco, Koley, Sandip, Samra, Nitzan, Rishal, Ida, Zhao, Qian, Sahoo, Pabitra K, Urisman, Anatoly, Marvaldi, Letizia, Oses-Prieto, Juan A, Forester, Craig, Gomes, Cynthia, Kalinski, Ashley L, Di Pizio, Agostina, Doron-Mandel, Ella, Perry, Rotem Ben-Tov, Koppel, Indrek, Twiss, Jeffery L, Burlingame, Alma L, and Fainzilber, Mike
- Subjects
Neurodegenerative ,Physical Injury - Accidents and Adverse Effects ,Neurosciences ,Genetics ,Underpinning research ,1.1 Normal biological development and functioning ,Neurological ,3' Untranslated Regions ,Animals ,Axons ,Cell Size ,Ganglia ,Spinal ,Mice ,Mice ,Inbred Strains ,Phosphoproteins ,Protein Biosynthesis ,RNA ,Messenger ,RNA-Binding Proteins ,Rats ,Rats ,Inbred BB ,Rats ,Sprague-Dawley ,Sciatic Nerve ,Signal Transduction ,TOR Serine-Threonine Kinases ,General Science & Technology - Abstract
How is protein synthesis initiated locally in neurons? We found that mTOR (mechanistic target of rapamycin) was activated and then up-regulated in injured axons, owing to local translation of mTOR messenger RNA (mRNA). This mRNA was transported into axons by the cell size-regulating RNA-binding protein nucleolin. Furthermore, mTOR controlled local translation in injured axons. This included regulation of its own translation and that of retrograde injury signaling molecules such as importin β1 and STAT3 (signal transducer and activator of transcription 3). Deletion of the mTOR 3' untranslated region (3'UTR) in mice reduced mTOR in axons and decreased local translation after nerve injury. Both pharmacological inhibition of mTOR in axons and deletion of the mTOR 3'UTR decreased proprioceptive neuronal survival after nerve injury. Thus, mRNA localization enables spatiotemporal control of mTOR pathways regulating local translation and long-range intracellular signaling.
- Published
- 2018
47. Revealing nascent proteomics in signaling pathways and cell differentiation
- Author
-
Forester, Craig M, Zhao, Qian, Phillips, Nancy J, Urisman, Anatoly, Chalkley, Robert J, Oses-Prieto, Juan A, Zhang, Li, Ruggero, Davide, and Burlingame, Alma L
- Subjects
Biotechnology ,Stem Cell Research ,Genetics ,Cell Differentiation ,Chromatography ,Liquid ,Drug Discovery ,Humans ,K562 Cells ,Protein Biosynthesis ,Proteome ,Proteomics ,Puromycin ,Signal Transduction ,TOR Serine-Threonine Kinases ,Tandem Mass Spectrometry ,proteomics ,translation ,mTOR ,erythropoiesis ,puromycin - Abstract
Regulation of gene expression at the level of protein synthesis is a crucial element in driving how the genetic landscape is expressed. However, we are still limited in technologies that can quantitatively capture the immediate proteomic changes that allow cells to respond to specific stimuli. Here, we present a method to capture and identify nascent proteomes in situ across different cell types without disturbing normal growth conditions, using O-propargyl-puromycin (OPP). Cell-permeable OPP rapidly labels nascent elongating polypeptides, which are subsequently conjugated to biotin-azide, using click chemistry, and captured with streptavidin beads, followed by digestion and analysis, using liquid chromatography-tandem mass spectrometry. Our technique of OPP-mediated identification (OPP-ID) allows detection of widespread proteomic changes within a short 2-hour pulse of OPP. We illustrate our technique by recapitulating alterations of proteomic networks induced by a potent mammalian target of rapamycin inhibitor, MLN128. In addition, by employing OPP-ID, we identify more than 2,100 proteins and uncover distinct protein networks underlying early erythroid progenitor and differentiation states not amenable to alternative approaches such as amino acid analog labeling. We present OPP-ID as a method to quantitatively identify nascent proteomes across an array of biological contexts while preserving the subtleties directing signaling in the native cellular environment.
- Published
- 2018
48. The Transcriptionally Permissive Chromatin State of Embryonic Stem Cells Is Acutely Tuned to Translational Output
- Author
-
Bulut-Karslioglu, Aydan, Macrae, Trisha A, Oses-Prieto, Juan A, Covarrubias, Sergio, Percharde, Michelle, Ku, Gregory, Diaz, Aaron, McManus, Michael T, Burlingame, Alma L, and Ramalho-Santos, Miguel
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Regenerative Medicine ,Stem Cell Research - Nonembryonic - Non-Human ,Genetics ,Human Genome ,Stem Cell Research ,Stem Cell Research - Embryonic - Non-Human ,1.1 Normal biological development and functioning ,Underpinning research ,Generic health relevance ,Animals ,Blastocyst ,Cell Differentiation ,Chromatin ,DNA Transposable Elements ,Embryonic Stem Cells ,Enhancer Elements ,Genetic ,Euchromatin ,Female ,Genome ,Histone Code ,Male ,Mice ,Models ,Biological ,Nuclear Proteins ,Protein Biosynthesis ,Protein Stability ,Proto-Oncogene Proteins c-myc ,RNA Interference ,TOR Serine-Threonine Kinases ,Transcription ,Genetic ,Chd1 ,blastocyst ,embryonic stem cells ,euchromatin ,hypertranscription ,mTOR ,permissive chromatin ,ribosome ,translation ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
A permissive chromatin environment coupled to hypertranscription drives the rapid proliferation of embryonic stem cells (ESCs) and peri-implantation embryos. We carried out a genome-wide screen to systematically dissect the regulation of the euchromatic state of ESCs. The results revealed that cellular growth pathways, most prominently translation, perpetuate the euchromatic state and hypertranscription of ESCs. Acute inhibition of translation rapidly depletes euchromatic marks in mouse ESCs and blastocysts, concurrent with delocalization of RNA polymerase II and reduction in nascent transcription. Translation inhibition promotes rewiring of chromatin accessibility, which decreases at a subset of active developmental enhancers and increases at histone genes and transposable elements. Proteome-scale analyses revealed that several euchromatin regulators are unstable proteins and continuously depend on a high translational output. We propose that this mechanistic interdependence of euchromatin, transcription, and translation sets the pace of proliferation at peri-implantation and may be employed by other stem/progenitor cells.
- Published
- 2018
49. Translatome Regulation in Neuronal Injury and Axon Regrowth
- Author
-
Rozenbaum, Meir, Rajman, Marek, Rishal, Ida, Koppel, Indrek, Koley, Sandip, Medzihradszky, Katalin F, Oses-Prieto, Juan A, Kawaguchi, Riki, Amieux, Paul S, Burlingame, Alma L, Coppola, Giovanni, and Fainzilber, Mike
- Subjects
Genetics ,Neurosciences ,Biotechnology ,Neurodegenerative ,Physical Injury - Accidents and Adverse Effects ,Underpinning research ,1.1 Normal biological development and functioning ,Neurological ,Animals ,Axons ,Cell Culture Techniques ,Ganglia ,Spinal ,Gene Expression Regulation ,Male ,Mice ,Mice ,Inbred C57BL ,Nerve Regeneration ,Neuronal Outgrowth ,Peripheral Nerve Injuries ,Protein Biosynthesis ,Proteomics ,Rats ,Rats ,Wistar ,Sensory Receptor Cells ,axon growth ,axon injury ,nerve regeneration ,translational regulation - Abstract
Transcriptional events leading to outgrowth of neuronal axons have been intensively studied, but the role of translational regulation in this process is not well understood. Here, we use translatome analyses by ribosome pull-down and protein synthesis characterization by metabolic isotopic labeling to study nerve injury and axon outgrowth proteomes in rodent dorsal root ganglia (DRGs) and sensory neurons. We identify over 1600 gene products that are primarily translationally regulated in DRG neurons after nerve injury, many of which contain a 5'UTR cytosine-enriched regulator of translation (CERT) motif, implicating the translation initiation factor Eif4e in the injury response. We further identified approximately 200 proteins that undergo robust de novo synthesis in the initial stages of axon growth. ApoE is one of the highly synthesized proteins in neurons, and its receptor binding inhibition or knockout affects axon outgrowth. These findings provide a resource for future analyses of the role of translational regulation in neuronal injury responses and axon extension.
- Published
- 2018
50. Hypoxia-ischemia modifies postsynaptic GluN2B-containing NMDA receptor complexes in the neonatal mouse brain
- Author
-
Lu, Fuxin, Shao, Guo, Wang, Yongqiang, Guan, Shenheng, Burlingame, Alma L, Liu, Xuemei, Liang, Xiao, Knox, Renatta, Ferriero, Donna M, and Jiang, Xiangning
- Subjects
Paediatrics ,Biomedical and Clinical Sciences ,Neurosciences ,Pediatric ,Brain Disorders ,1.1 Normal biological development and functioning ,Underpinning research ,Neurological ,Animals ,Animals ,Newborn ,Brain ,Brain Chemistry ,Female ,Hypoxia-Ischemia ,Brain ,Immunoprecipitation ,Male ,Mice ,Mice ,Inbred C57BL ,Neural Pathways ,Neurons ,Pregnancy ,Primary Cell Culture ,Protein Kinases ,Proteomics ,Receptors ,N-Methyl-D-Aspartate ,Receptors ,Neurotransmitter ,Development ,Hypoxia/ischemia ,NMDA receptors ,Clinical Sciences ,Psychology ,Neurology & Neurosurgery ,Biological psychology - Abstract
The N-methyl-d-aspartate-type glutamate receptor (NMDAR)-associated multiprotein complexes are indispensable for synaptic plasticity and cognitive functions. While purification and proteomic analyses of these signaling complexes have been performed in adult rodent and human brain, much less is known about the protein composition of NMDAR complexes in the developing brain and their modifications by neonatal hypoxic-ischemic (HI) brain injury. In this study, the postsynaptic density proteins were prepared from postnatal day 9 naïve, sham-operated and HI-injured mouse cortex. The GluN2B-containing NMDAR complexes were purified by immunoprecipitation with a mouse GluN2B antibody and subjected to mass spectrometry analysis for determination of the GluN2B binding partners. A total of 71 proteins of different functional categories were identified from the naïve animals as native GluN2B-interacting partners in the developing mouse brain. Neonatal HI reshaped the postsynaptic GluN2B interactome by recruiting new proteins, including multiple kinases, into the complexes; and modifying the existing associations within 1h of reperfusion. The early responses of postsynaptic NMDAR complexes and their related signaling networks may contribute to molecular processes leading to cell survival or death, brain damage and/or neurological disorders in term infants with neonatal encephalopathy.
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.