141,390 results on '"HEK293 Cells"'
Search Results
2. Enhancers and genome conformation provide complex transcriptional control of a herpesviral gene
- Author
-
Morgens, David W, Gulyas, Leah, Mao, Xiaowen, Rivera-Madera, Alejandro, Souza, Annabelle S, and Glaunsinger, Britt A
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Genetics ,Biotechnology ,Emerging Infectious Diseases ,Human Genome ,Infectious Diseases ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Generic health relevance ,Herpesvirus 8 ,Human ,Humans ,Gene Expression Regulation ,Viral ,Genome ,Viral ,Transcription ,Genetic ,Promoter Regions ,Genetic ,Enhancer Elements ,Genetic ,Viral Proteins ,HEK293 Cells ,Capture Hi-C ,CRISPR Interference ,Gene Regulation ,Herpesvirus ,KSHV ,Biochemistry and Cell Biology ,Other Biological Sciences ,Bioinformatics ,Biochemistry and cell biology - Abstract
Complex transcriptional control is a conserved feature of both eukaryotes and the viruses that infect them. Despite viral genomes being smaller and more gene dense than their hosts, we generally lack a sense of scope for the features governing the transcriptional output of individual viral genes. Even having a seemingly simple expression pattern does not imply that a gene's underlying regulation is straightforward. Here, we illustrate this by combining high-density functional genomics, expression profiling, and viral-specific chromosome conformation capture to define with unprecedented detail the transcriptional regulation of a single gene from Kaposi's sarcoma-associated herpesvirus (KSHV). We used as our model KSHV ORF68 - which has simple, early expression kinetics and is essential for viral genome packaging. We first identified seven cis-regulatory regions involved in ORF68 expression by densely tiling the ~154 kb KSHV genome with dCas9 fused to a transcriptional repressor domain (CRISPRi). A parallel Cas9 nuclease screen indicated that three of these regions act as promoters of genes that regulate ORF68. RNA expression profiling demonstrated that three more of these regions act by either repressing or enhancing other distal viral genes involved in ORF68 transcriptional regulation. Finally, we tracked how the 3D structure of the viral genome changes during its lifecycle, revealing that these enhancing regulatory elements are physically closer to their targets when active, and that disrupting some elements caused large-scale changes to the 3D genome. These data enable us to construct a complete model revealing that the mechanistic diversity of this essential regulatory circuit matches that of human genes.
- Published
- 2025
3. Activity-assembled nBAF complex mediates rapid immediate early gene transcription by regulating RNA polymerase II productive elongation.
- Author
-
Cornejo, Karen, Venegas, Andie, Sono, Morgan, Door, Madeline, Gutierrez-Ruiz, Brenda, Karabedian, Lucy, Nandi, Supratik, Hadisurya, Marco, Tao, W, Dykhuizen, Emily, and Saha, Ramendra
- Subjects
Arc ,BD98 ,CP: Molecular biology ,CP: Neuroscience ,RNA Pol II ,RNA polymerase II ,SWI/SNF ,immediate early gene ,nBAF ,neuron ,productive elongation ,transcription ,RNA Polymerase II ,Transcription Factors ,Humans ,Animals ,Nuclear Proteins ,Transcription ,Genetic ,Genes ,Immediate-Early ,Transcription Elongation ,Genetic ,Mice ,HEK293 Cells ,Promoter Regions ,Genetic ,Neurons - Abstract
Signal-dependent RNA polymerase II (RNA Pol II) productive elongation is an integral component of gene transcription, including that of immediate early genes (IEGs) induced by neuronal activity. However, it remains unclear how productively elongating RNA Pol II overcomes nucleosomal barriers. Using RNAi, three degraders, and several small-molecule inhibitors, we show that the mammalian switch/sucrose non-fermentable (SWI/SNF) complex of neurons (neuronal BRG1/BRM-associated factor or nBAF) is required for activity-induced transcription of neuronal IEGs, including Arc. The nBAF complex facilitates promoter-proximal RNA Pol II pausing and signal-dependent RNA Pol II recruitment (loading) and, importantly, mediates productive elongation in the gene body via interaction with the elongation complex and elongation-competent RNA Pol II. Mechanistically, RNA Pol II elongation is mediated by activity-induced nBAF assembly (especially ARID1A recruitment) and its ATPase activity. Together, our data demonstrate that the nBAF complex regulates several aspects of RNA Pol II transcription and reveal mechanisms underlying activity-induced RNA Pol II elongation. These findings may offer insights into human maladies etiologically associated with mutational interdiction of BAF functions.
- Published
- 2024
4. SPOP-mediated RIPK3 destabilization desensitizes LPS/sMAC/zVAD-induced necroptotic cell death.
- Author
-
Lee, Ga-Eun, Bang, Geul, Byun, Jiin, Chen, Weidong, Jeung, Dohyun, Cho, Hana, Lee, Joo, Kang, Han, Lee, Hye, Kim, Jin, Kim, Kwang, Wu, Juan, Nam, Soo-Bin, Kwon, Young, Lee, Cheol-Jung, and Cho, Yong-Yeon
- Subjects
Kinases ,Necroptosis ,RIPK3 ,SPOP ,Ubiquitination ,Receptor-Interacting Protein Serine-Threonine Kinases ,Humans ,Nuclear Proteins ,Repressor Proteins ,Ubiquitination ,Phosphorylation ,Lipopolysaccharides ,Colonic Neoplasms ,Necroptosis ,HEK293 Cells ,Cell Line ,Tumor ,Cell Death ,Protein Stability ,Protein Binding - Abstract
RIPK1/RIPK3-MLKL signaling molecules are fundamental in initiating necroptotic cell death, but their roles in the development of colon cancer are unclear. This study reports that RIPK3 interacted with SPOP, a component of the E3 ligase within the Cul3 complex. This interaction leads to K48-linked ubiquitination and subsequent proteasomal degradation of RIPK3. Two distinct degron motifs, PETST and SPTST, were identified within the linker domain of RIPK3 for SPOP. RIPK3 phosphorylations at Thr403 by PIM2 and at Thr412/Ser413 by ERK2 are essential to facilitate its interaction with SPOP. Computational docking studies and immunoprecipitation analyses showed that these PIM2 and ERK2 phosphorylations bolster the stability of the RIPK3-SPOP interaction. In particular, mutations of RIPK3 at the degron motifs extended the half-life of RIPK3 by preventing its phosphorylation and subsequent ubiquitination. The deletion of SPOP, which led to increased stability of the RIPK3 protein, intensified LPS/sMAC/zVAD-induced necroptotic cell death in colon cancer cells. These findings underscore the critical role of the SPOP-mediated RIPK3 stability regulation pathway in controlling necroptotic cell death.
- Published
- 2024
5. A CRISPR-Cas9 knockout screening identifies IRF2 as a key driver of OAS3/RNase L-mediated RNA decay during viral infection
- Author
-
Oh, Sunwoo, Santiago, Gisselle, Manjunath, Lavanya, Li, Junyi, Bouin, Alexis, Semler, Bert L, and Buisson, Rémi
- Subjects
Medical Microbiology ,Biomedical and Clinical Sciences ,Biological Sciences ,Emerging Infectious Diseases ,Genetics ,Infectious Diseases ,2.1 Biological and endogenous factors ,1.1 Normal biological development and functioning ,Infection ,CRISPR-Cas Systems ,Humans ,Interferon Regulatory Factor-2 ,Endoribonucleases ,2' ,5'-Oligoadenylate Synthetase ,RNA Stability ,Virus Diseases ,Gene Knockout Techniques ,RNA ,Viral ,HEK293 Cells ,Virus Replication ,STAT2 Transcription Factor ,innate immunity ,RNase ,RNA decay ,CRISPR screen ,virus ,RNase L - Abstract
OAS-RNase L is a double-stranded RNA-induced antiviral pathway triggered in response to diverse viral infections. Upon activation, OAS-RNase L suppresses virus replication by promoting the decay of host and viral RNAs and inducing translational shutdown. However, whether OASs and RNase L are the only factors involved in this pathway remains unclear. Here, we develop CRISPR-Translate, a FACS-based genome-wide CRISPR-Cas9 knockout screening method that uses translation levels as a readout and identifies IRF2 as a key regulator of OAS3. Mechanistically, we demonstrate that IRF2 promotes basal expression of OAS3 in unstressed cells, allowing a rapid activation of RNase L following viral infection. Furthermore, IRF2 works in concert with the interferon response through STAT2 to further enhance OAS3 expression. We propose that IRF2-induced RNase L is critical in enabling cells to mount a rapid antiviral response immediately after viral infection, serving as the initial line of defense. This rapid response provides host cells the necessary time to activate additional antiviral signaling pathways, forming secondary defense waves.
- Published
- 2024
6. Ketamine and Major Ketamine Metabolites Function as Allosteric Modulators of Opioid Receptors
- Author
-
Gomes, Ivone, Gupta, Achla, Margolis, Elyssa B, Fricker, Lloyd D, and Devi, Lakshmi A
- Subjects
Pharmacology and Pharmaceutical Sciences ,Biomedical and Clinical Sciences ,Neurosciences ,Drug Abuse (NIDA only) ,Opioids ,Brain Disorders ,Substance Misuse ,1.1 Normal biological development and functioning ,5.1 Pharmaceuticals ,Neurological ,Generic health relevance ,Ketamine ,Animals ,Allosteric Regulation ,Receptors ,Opioid ,Humans ,Mice ,Rats ,Male ,Rats ,Sprague-Dawley ,HEK293 Cells ,Cricetulus ,CHO Cells ,Biochemistry and Cell Biology ,Pharmacology & Pharmacy ,Biochemistry and cell biology ,Pharmacology and pharmaceutical sciences - Abstract
Ketamine is a glutamate receptor antagonist that was developed over 50 years ago as an anesthetic agent. At subanesthetic doses, ketamine and some metabolites are analgesics and fast-acting antidepressants, presumably through targets other than glutamate receptors. We tested ketamine and its metabolites for activity as allosteric modulators of opioid receptors expressed as recombinant receptors in heterologous systems and with native receptors in rodent brain; signaling was examined by measuring GTP binding, β-arrestin recruitment, MAPK activation, and neurotransmitter release. Although micromolar concentrations of ketamine alone had weak agonist activity at μ opioid receptors, the combination of submicromolar concentrations of ketamine with endogenous opioid peptides produced robust synergistic responses with statistically significant increases in efficacies. All three opioid receptors (μ, δ, and κ) showed synergism with submicromolar concentrations of ketamine and either methionine-enkephalin (Met-enk), leucine-enkephalin (Leu-enk), and/or dynorphin A17 (Dyn A17), albeit the extent of synergy was variable between receptors and peptides. S-ketamine exhibited higher modulatory effects compared with R-ketamine or racemic ketamine, with ∼100% increase in efficacy. Importantly, the ketamine metabolite 6-hydroxynorketamine showed robust allosteric modulatory activity at μ opioid receptors; this metabolite is known to have analgesic and antidepressant activity but does not bind to glutamate receptors. Ketamine enhanced potency and efficacy of Met-enkephalin signaling both in mouse midbrain membranes and in rat ventral tegmental area neurons as determined by electrophysiology recordings in brain slices. Taken together, these findings support the hypothesis that some of the therapeutic effects of ketamine and its metabolites are mediated by directly engaging the endogenous opioid system. SIGNIFICANCE STATEMENT: This study found that ketamine and its major biologically active metabolites function as potent allosteric modulators of μ, δ, and κ opioid receptors, with submicromolar concentrations of these compounds synergizing with endogenous opioid peptides, such as enkephalin and dynorphin. This allosteric activity may contribute to ketamine's therapeutic effectiveness for treating acute and chronic pain and as a fast-acting antidepressant drug.
- Published
- 2024
7. Distinct pathways for evolution of enhanced receptor binding and cell entry in SARS-like bat coronaviruses.
- Author
-
Tse, Alexandra, Acreman, Cory, Ricardo-Lax, Inna, Berrigan, Jacob, Lasso, Gorka, Balogun, Toheeb, Kearns, Fiona, Casalino, Lorenzo, McClain, Georgia, Chandran, Amartya, Lemeunier, Charlotte, Amaro, Rommie, Rice, Charles, Jangra, Rohit, McLellan, Jason, Chandran, Kartik, and Miller, Emily
- Subjects
Animals ,Chiroptera ,Virus Internalization ,Spike Glycoprotein ,Coronavirus ,Humans ,Angiotensin-Converting Enzyme 2 ,Receptors ,Virus ,Severe acute respiratory syndrome-related coronavirus ,SARS-CoV-2 ,HEK293 Cells ,Coronavirus Infections - Abstract
Understanding the zoonotic risks posed by bat coronaviruses (CoVs) is critical for pandemic preparedness. Herein, we generated recombinant vesicular stomatitis viruses (rVSVs) bearing spikes from divergent bat CoVs to investigate their cell entry mechanisms. Unexpectedly, the successful recovery of rVSVs bearing the spike from SHC014-CoV, a SARS-like bat CoV, was associated with the acquisition of a novel substitution in the S2 fusion peptide-proximal region (FPPR). This substitution enhanced viral entry in both VSV and coronavirus contexts by increasing the availability of the spike receptor-binding domain to recognize its cellular receptor, ACE2. A second substitution in the S1 N-terminal domain, uncovered through the rescue and serial passage of a virus bearing the FPPR substitution, further enhanced spike:ACE2 interaction and viral entry. Our findings identify genetic pathways for adaptation by bat CoVs during spillover and host-to-host transmission, fitness trade-offs inherent to these pathways, and potential Achilles heels that could be targeted with countermeasures.
- Published
- 2024
8. Orientation-dependent CD45 inhibition with viral and engineered ligands.
- Author
-
Borowska, Marta, Liu, Liu, Caveney, Nathanael, Jude, Kevin, Kim, Won-Ju, Masubuchi, Takeya, Hui, Enfu, Majzner, Robbie, and Garcia, K
- Subjects
Leukocyte Common Antigens ,Humans ,Ligands ,T-Lymphocytes ,Viral Proteins ,Adenoviridae ,Lymphocyte Activation ,HEK293 Cells ,Cryoelectron Microscopy - Abstract
CD45 is a cell surface phosphatase that shapes the T cell receptor signaling threshold but does not have a known ligand. A family of adenovirus proteins, including E3/49K, exploits CD45 to evade immunity by binding to the extracellular domain of CD45, resulting in the suppression of T cell signaling. We determined the cryo-EM structure of this complex and found that the E3/49K protein is composed of three immunoglobulin domains assembled as beads on a string that compel CD45 into a closely abutted dimer by cross-linking the CD45 D3 domain, leading to steric inhibition of its intracellular phosphatase activity. Inspired by the E3/49K mechanism, we engineered CD45 surrogate ligands that can fine-tune T cell activation by dimerizing CD45 into different orientations and proximities. The adenovirus E3/49K protein has taught us that, despite a lack of a known ligand, CD45 activity can be modulated by extracellular dimerizing ligands that perturb its phosphatase activity and alter T cell responses.
- Published
- 2024
9. Structural basis of μ-opioid receptor targeting by a nanobody antagonist.
- Author
-
Yu, Jun, Kumar, Amit, Zhang, Xuefeng, Martin, Charlotte, Van Holsbeeck, Kevin, Raia, Pierre, Koehl, Antoine, Laeremans, Toon, Steyaert, Jan, Manglik, Aashish, Ballet, Steven, Boland, Andreas, and Stoeber, Miriam
- Subjects
Receptors ,Opioid ,mu ,Single-Domain Antibodies ,Humans ,Cryoelectron Microscopy ,Ligands ,HEK293 Cells ,Animals ,Protein Binding ,Binding Sites ,Models ,Molecular ,Analgesics ,Opioid ,Peptides ,Cyclic - Abstract
The μ-opioid receptor (μOR), a prototypical G protein-coupled receptor (GPCR), is the target of opioid analgesics such as morphine and fentanyl. Due to the severe side effects of current opioid drugs, there is considerable interest in developing novel modulators of μOR function. Most GPCR ligands today are small molecules, however biologics, including antibodies and nanobodies, represent alternative therapeutics with clear advantages such as affinity and target selectivity. Here, we describe the nanobody NbE, which selectively binds to the μOR and acts as an antagonist. We functionally characterize NbE as an extracellular and genetically encoded μOR ligand and uncover the molecular basis for μOR antagonism by determining the cryo-EM structure of the NbE-μOR complex. NbE displays a unique ligand binding mode and achieves μOR selectivity by interactions with the orthosteric pocket and extracellular receptor loops. Based on a β-hairpin loop formed by NbE that deeply protrudes into the μOR, we design linear and cyclic peptide analogs that recapitulate NbEs antagonism. The work illustrates the potential of nanobodies to uniquely engage with GPCRs and describes lower molecular weight μOR ligands that can serve as a basis for therapeutic developments.
- Published
- 2024
10. Isoform-specific C-terminal phosphorylation drives autoinhibition of Casein kinase 1.
- Author
-
Harold, Rachel, Tulsian, Nikhil, Narasimamurthy, Rajesh, Yaitanes, Noelle, Ayala Hernandez, Maria, Lee, Hsiau-Wei, Crosby, Priya, Tripathi, Sarvind, Virshup, David, and Partch, Carrie
- Subjects
circadian rhythms ,intrinsically disordered proteins ,kinase ,Phosphorylation ,Humans ,Casein Kinase Idelta ,Circadian Rhythm ,Animals ,Casein Kinase I ,HEK293 Cells ,Mice ,Protein Domains ,Mutation - Abstract
Casein kinase 1δ (CK1δ) controls essential biological processes including circadian rhythms and wingless-related integration site (Wnt) signaling, but how its activity is regulated is not well understood. CK1δ is inhibited by autophosphorylation of its intrinsically disordered C-terminal tail. Two CK1 splice variants, δ1 and δ2, are known to have very different effects on circadian rhythms. These variants differ only in the last 16 residues of the tail, referred to as the extreme C termini (XCT), but with marked changes in potential phosphorylation sites. Here, we test whether the XCT of these variants have different effects in autoinhibition of the kinase. Using NMR and hydrogen/deuterium exchange mass spectrometry, we show that the δ1 XCT is preferentially phosphorylated by the kinase and the δ1 tail makes more extensive interactions across the kinase domain. Mutation of δ1-specific XCT phosphorylation sites increases kinase activity both in vitro and in cells and leads to changes in the circadian period, similar to what is reported in vivo. Mechanistically, loss of the phosphorylation sites in XCT disrupts tail interaction with the kinase domain. δ1 autoinhibition relies on conserved anion-binding sites around the CK1 active site, demonstrating a common mode of product inhibition of CK1δ. These findings demonstrate how a phosphorylation cycle controls the activity of this essential kinase.
- Published
- 2024
11. A genome-wide screen links peroxisome regulation with Wnt signaling through RNF146 and TNKS/2
- Author
-
Vu, Jonathan T, Tavasoli, Katherine U, Sheedy, Connor J, Chowdhury, Soham P, Mandjikian, Lori, Bacal, Julien, Morrissey, Meghan A, Richardson, Chris D, and Gardner, Brooke M
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Genetics ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Generic health relevance ,Peroxisomes ,Ubiquitin-Protein Ligases ,Humans ,Wnt Signaling Pathway ,Axin Protein ,Membrane Proteins ,beta Catenin ,HEK293 Cells ,Protein Transport ,CRISPR-Cas Systems ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
Peroxisomes are membrane-bound organelles harboring metabolic enzymes. In humans, peroxisomes are required for normal development, yet the genes regulating peroxisome function remain unclear. We performed a genome-wide CRISPRi screen to identify novel factors involved in peroxisomal homeostasis. We found that inhibition of RNF146, an E3 ligase activated by poly(ADP-ribose), reduced the import of proteins into peroxisomes. RNF146-mediated loss of peroxisome import depended on the stabilization and activity of the poly(ADP-ribose) polymerases TNKS and TNKS2, which bind the peroxisomal membrane protein PEX14. We propose that RNF146 and TNKS/2 regulate peroxisome import efficiency by PARsylation of proteins at the peroxisome membrane. Interestingly, we found that the loss of peroxisomes increased TNKS/2 and RNF146-dependent degradation of non-peroxisomal substrates, including the β-catenin destruction complex component AXIN1, which was sufficient to alter the amplitude of β-catenin transcription. Together, these observations not only suggest previously undescribed roles for RNF146 in peroxisomal regulation but also a novel role in bridging peroxisome function with Wnt/β-catenin signaling during development.
- Published
- 2024
12. Large-scale map of RNA-binding protein interactomes across the mRNA life cycle
- Author
-
Street, Lena A, Rothamel, Katherine L, Brannan, Kristopher W, Jin, Wenhao, Bokor, Benjamin J, Dong, Kevin, Rhine, Kevin, Madrigal, Assael, Al-Azzam, Norah, Kim, Jenny Kim, Ma, Yanzhe, Gorhe, Darvesh, Abdou, Ahmed, Wolin, Erica, Mizrahi, Orel, Ahdout, Joshua, Mujumdar, Mayuresh, Doron-Mandel, Ella, Jovanovic, Marko, and Yeo, Gene W
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Genetics ,1.1 Normal biological development and functioning ,Generic health relevance ,Neurological ,Humans ,RNA ,Messenger ,RNA-Binding Proteins ,Protein Interaction Maps ,Protein Binding ,HeLa Cells ,Protein Interaction Mapping ,Ribonucleoproteins ,Small Nuclear ,HEK293 Cells ,Mass Spectrometry ,RNA Splicing ,Hela Cells ,ERH ,IP-MS ,RBP ,RNA-binding proteins ,RNA-dependent protein interactions ,SEC-MS ,SNRNP200 ,interactome ,mRNA life-cycle ,protein-protein interactions ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
mRNAs interact with RNA-binding proteins (RBPs) throughout their processing and maturation. While efforts have assigned RBPs to RNA substrates, less exploration has leveraged protein-protein interactions (PPIs) to study proteins in mRNA life-cycle stages. We generated an RNA-aware, RBP-centric PPI map across the mRNA life cycle in human cells by immunopurification-mass spectrometry (IP-MS) of ∼100 endogenous RBPs with and without RNase, augmented by size exclusion chromatography-mass spectrometry (SEC-MS). We identify 8,742 known and 20,802 unreported interactions between 1,125 proteins and determine that 73% of the IP-MS-identified interactions are RNA regulated. Our interactome links many proteins, some with unknown functions, to specific mRNA life-cycle stages, with nearly half associated with multiple stages. We demonstrate the value of this resource by characterizing the splicing and export functions of enhancer of rudimentary homolog (ERH), and by showing that small nuclear ribonucleoprotein U5 subunit 200 (SNRNP200) interacts with stress granule proteins and binds cytoplasmic RNA differently during stress.
- Published
- 2024
13. Molecular mechanism of contactin 2 homophilic interaction
- Author
-
Fan, Shanghua, Liu, Jianfang, Chofflet, Nicolas, Bailey, Aaron O, Russell, William K, Zhang, Ziqi, Takahashi, Hideto, Ren, Gang, and Rudenko, Gabby
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Neurological ,Humans ,Protein Binding ,Protein Multimerization ,Cryoelectron Microscopy ,Contactin 2 ,Models ,Molecular ,Binding Sites ,HEK293 Cells ,adhesion molecule ,contactins ,cross-linking mass spectrometry ,cryo-EM ,molecular recognition mechanism ,molecular specificity ,neuronal guidance molecule ,protein structure ,protein:protein interaction ,single-particle analysis ,Chemical Sciences ,Information and Computing Sciences ,Biophysics ,Biological sciences ,Chemical sciences - Abstract
Contactin 2 (CNTN2) is a cell adhesion molecule involved in axon guidance, neuronal migration, and fasciculation. The ectodomains of CNTN1-CNTN6 are composed of six Ig domains (Ig1-Ig6) and four FN domains. Here, we show that CNTN2 forms transient homophilic interactions (KD ∼200 nM). Cryo-EM structures of full-length CNTN2 and CNTN2_Ig1-Ig6 reveal a T-shaped homodimer formed by intertwined, parallel monomers. Unexpectedly, the horseshoe-shaped Ig1-Ig4 headpieces extend their Ig2-Ig3 tips outwards on either side of the homodimer, while Ig4, Ig5, Ig6, and the FN domains form a central stalk. Cross-linking mass spectrometry and cell-based binding assays confirm the 3D assembly of the CNTN2 homodimer. The interface mediating homodimer formation differs between CNTNs, as do the homophilic versus heterophilic interaction mechanisms. The CNTN family thus encodes a versatile molecular platform that supports a very diverse portfolio of protein interactions and that can be leveraged to strategically guide neural circuit development.
- Published
- 2024
14. Modulating the unfolded protein response with ISRIB mitigates cisplatin ototoxicity.
- Author
-
Li, Jiang, Rouse, Stephanie, Matthews, Ian, Park, Yesai, Eltawil, Yasmin, Sherr, Elliott, and Chan, Dylan
- Subjects
Cisplatin ,Endoplasmic reticulum stress ,Ototoxicity ,Unfolded protein response ,Cisplatin ,Unfolded Protein Response ,Animals ,Ototoxicity ,Humans ,Mice ,Endoplasmic Reticulum Stress ,HEK293 Cells ,Antineoplastic Agents ,Apoptosis ,Cochlea ,Hair Cells ,Auditory ,eIF-2 Kinase - Abstract
Cisplatin is a commonly used chemotherapy agent with a nearly universal side effect of sensorineural hearing loss. The cellular mechanisms underlying cisplatin ototoxicity are poorly understood. Efforts in drug development to prevent or reverse cisplatin ototoxicity have largely focused on pathways of oxidative stress and apoptosis. An effective treatment for cisplatin ototoxicity, sodium thiosulfate (STS), while beneficial when used in standard risk hepatoblastoma, is associated with reduced survival in disseminated pediatric malignancy, highlighting the need for more specific drugs without potential tumor protective effects. The unfolded protein response (UPR) and endoplasmic reticulum (ER) stress pathways have been shown to be involved in the pathogenesis of noise-induced hearing loss and cochlear synaptopathy in vivo, and these pathways have been implicated broadly in cisplatin cytotoxicity. This study sought to determine whether the UPR can be targeted to prevent cisplatin ototoxicity. Neonatal cochlear cultures and HEK cells were exposed to cisplatin, and UPR marker gene expression and cell death measured. Treatment with ISRIB (Integrated Stress Response InhIBitor), a drug that activates eif2B and downregulates the pro-apoptotic PERK/CHOP pathway of the UPR, was tested for its ability to reduce apoptosis in HEK cells, hair-cell death in cochlear cultures, and hearing loss using an in vivo mouse model of cisplatin ototoxicity. Finally, to evaluate whether ISRIB might interfere with cisplatin chemoeffectiveness, we tested it in head and neck squamous cell carcinoma (HNSCC) cell-based assays of cisplatin cytotoxicity. Cisplatin exhibited a biphasic, non-linear dose-response of cell death and apoptosis that correlated with different patterns of UPR marker gene expression in HEK cells and cochlear cultures. ISRIB treatment protected against cisplatin-induced hearing loss and hair-cell death, but did not impact cisplatins cytotoxic effects on HNSCC cell viability, unlike STS. These findings demonstrate that targeting the pro-apoptotic PERK/CHOP pathway with ISRIB can mitigate cisplatin ototoxicity without reducing anti-cancer cell effects, suggesting that this may be a viable strategy for drug development.
- Published
- 2024
15. Systematic identification of post-transcriptional regulatory modules.
- Author
-
Khoroshkin, Matvei, Buyan, Andrey, Dodel, Martin, Navickas, Albertas, Yu, Johnny, Trejo, Fathima, Doty, Anthony, Baratam, Rithvik, Zhou, Shaopu, Lee, Sean, Joshi, Tanvi, Garcia, Kristle, Choi, Benedict, Miglani, Sohit, Subramanyam, Vishvak, Modi, Hailey, Carpenter, Christopher, Markett, Daniel, Corces, M, Mardakheh, Faraz, Kulakovskiy, Ivan, and Goodarzi, Hani
- Subjects
Humans ,RNA-Binding Proteins ,RNA ,Messenger ,Transcriptome ,RNA Processing ,Post-Transcriptional ,Gene Expression Regulation ,HEK293 Cells ,Single-Cell Analysis ,Gene Regulatory Networks ,Regulon - Abstract
In our cells, a limited number of RNA binding proteins (RBPs) are responsible for all aspects of RNA metabolism across the entire transcriptome. To accomplish this, RBPs form regulatory units that act on specific target regulons. However, the landscape of RBP combinatorial interactions remains poorly explored. Here, we perform a systematic annotation of RBP combinatorial interactions via multimodal data integration. We build a large-scale map of RBP protein neighborhoods by generating in vivo proximity-dependent biotinylation datasets of 50 human RBPs. In parallel, we use CRISPR interference with single-cell readout to capture transcriptomic changes upon RBP knockdowns. By combining these physical and functional interaction readouts, along with the atlas of RBP mRNA targets from eCLIP assays, we generate an integrated map of functional RBP interactions. We then use this map to match RBPs to their context-specific functions and validate the predicted functions biochemically for four RBPs. This study provides a detailed map of RBP interactions and deconvolves them into distinct regulatory modules with annotated functions and target regulons. This multimodal and integrative framework provides a principled approach for studying post-transcriptional regulatory processes and enriches our understanding of their underlying mechanisms.
- Published
- 2024
16. Light-gated integrator for highlighting kinase activity in living cells.
- Author
-
Lin, Wei, Phatarphekar, Abhishek, Zhong, Yanghao, Liu, Longwei, Kwon, Hyung-Bae, Gerwick, William, Wang, Yingxiao, Mehta, Sohum, and Zhang, Jin
- Subjects
Humans ,Cyclic AMP-Dependent Protein Kinases ,HEK293 Cells ,Signal Transduction ,Light ,Biosensing Techniques - Abstract
Protein kinases are key signaling nodes that regulate fundamental biological and disease processes. Illuminating kinase signaling from multiple angles can provide deeper insights into disease mechanisms and improve therapeutic targeting. While fluorescent biosensors are powerful tools for visualizing live-cell kinase activity dynamics in real time, new molecular tools are needed that enable recording of transient signaling activities for post hoc analysis and targeted manipulation. Here, we develop a light-gated kinase activity coupled transcriptional integrator (KINACT) that converts dynamic kinase signals into permanent fluorescent marks. KINACT enables robust monitoring of kinase activity across scales, accurately recording subcellular PKA activity, highlighting PKA activity distribution in 3D cultures, and identifying PKA activators and inhibitors in high-throughput screens. We further leverage the ability of KINACT to drive signaling effector expression to allow feedback manipulation of the balance of GαsR201C-induced PKA and ERK activation and dissect the mechanisms of oncogenic G protein signaling.
- Published
- 2024
17. Structural basis of TRPV1 modulation by endogenous bioactive lipids
- Author
-
Arnold, William R, Mancino, Adamo, Moss, Frank R, Frost, Adam, Julius, David, and Cheng, Yifan
- Subjects
Biomedical and Clinical Sciences ,Neurosciences ,Pain Research ,Chronic Pain ,5.1 Pharmaceuticals ,TRPV Cation Channels ,Animals ,Rats ,Lysophospholipids ,Binding Sites ,Protein Binding ,Models ,Molecular ,Humans ,Phosphatidylinositols ,Protein Conformation ,HEK293 Cells ,Chemical Sciences ,Biological Sciences ,Medical and Health Sciences ,Biophysics ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences ,Chemical sciences - Abstract
TRP ion channels are modulated by phosphoinositide lipids, but the underlying structural mechanisms remain unclear. The capsaicin- and heat-activated receptor, TRPV1, has served as a model for deciphering lipid modulation, which is relevant to understanding how pro-algesic agents enhance channel activity in the setting of inflammatory pain. Identification of a pocket within the TRPV1 transmembrane core has provided initial clues as to how phosphoinositide lipids bind to and regulate the channel. Here we show that this regulatory pocket in rat TRPV1 can accommodate diverse lipid species, including the inflammatory lipid lysophosphatidic acid, whose actions are determined by their specific modes of binding. Furthermore, we show that an empty-pocket channel lacking an endogenous phosphoinositide lipid assumes an agonist-like state, even at low temperature, substantiating the concept that phosphoinositide lipids serve as negative TRPV1 modulators whose ejection from the binding pocket is a critical step toward activation by thermal or chemical stimuli.
- Published
- 2024
18. Large-scale evaluation of the ability of RNA-binding proteins to activate exon inclusion
- Author
-
Schmok, Jonathan C, Jain, Manya, Street, Lena A, Tankka, Alex T, Schafer, Danielle, Her, Hsuan-Lin, Elmsaouri, Sara, Gosztyla, Maya L, Boyle, Evan A, Jagannatha, Pratibha, Luo, En-Ching, Kwon, Ester J, Jovanovic, Marko, and Yeo, Gene W
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Genetics ,Humans ,Exons ,RNA-Binding Proteins ,Alternative Splicing ,HEK293 Cells - Abstract
RNA-binding proteins (RBPs) modulate alternative splicing outcomes to determine isoform expression and cellular survival. To identify RBPs that directly drive alternative exon inclusion, we developed tethered function luciferase-based splicing reporters that provide rapid, scalable and robust readouts of exon inclusion changes and used these to evaluate 718 human RBPs. We performed enhanced cross-linking immunoprecipitation, RNA sequencing and affinity purification-mass spectrometry to investigate a subset of candidates with no prior association with splicing. Integrative analysis of these assays indicates surprising roles for TRNAU1AP, SCAF8 and RTCA in the modulation of hundreds of endogenous splicing events. We also leveraged our tethering assays and top candidates to identify potent and compact exon inclusion activation domains for splicing modulation applications. Using these identified domains, we engineered programmable fusion proteins that outperform current artificial splicing factors at manipulating inclusion of reporter and endogenous exons. This tethering approach characterizes the ability of RBPs to induce exon inclusion and yields new molecular parts for programmable splicing control.
- Published
- 2024
19. Multi-modal contrastive learning of subcellular organization using DICE.
- Author
-
Nasser, Rami, Schaffer, Leah, Ideker, Trey, and Sharan, Roded
- Subjects
Humans ,HEK293 Cells ,Computational Biology ,Protein Interaction Mapping ,Proteins ,Unsupervised Machine Learning - Abstract
The data deluge in biology calls for computational approaches that can integrate multiple datasets of different types to build a holistic view of biological processes or structures of interest. An emerging paradigm in this domain is the unsupervised learning of data embeddings that can be used for downstream clustering and classification tasks. While such approaches for integrating data of similar types are becoming common, there is scarcer work on consolidating different data modalities such as network and image information. Here, we introduce DICE (Data Integration through Contrastive Embedding), a contrastive learning model for multi-modal data integration. We apply this model to study the subcellular organization of proteins by integrating protein-protein interaction data and protein image data measured in HEK293 cells. We demonstrate the advantage of data integration over any single modality and show that our framework outperforms previous integration approaches. Availability: https://github.com/raminass/protein-contrastive Contact: raminass@gmail.com.
- Published
- 2024
20. Spatial organization of adenylyl cyclase and its impact on dopamine signaling in neurons
- Author
-
Ripoll, Léa, Li, Yong, Dessauer, Carmen W, and von Zastrow, Mark
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Neurosciences ,Adenylyl Cyclases ,Animals ,Signal Transduction ,Dopamine ,Neurons ,Cyclic AMP ,Cyclic AMP-Dependent Protein Kinases ,Endosomes ,Cell Membrane ,Mice ,Corpus Striatum ,Receptors ,Dopamine ,Golgi Apparatus ,Cell Nucleus ,Humans ,HEK293 Cells - Abstract
The cAMP cascade is increasingly recognized to transduce physiological effects locally through spatially limited cAMP gradients. However, little is known about how adenylyl cyclase enzymes that initiate cAMP gradients are localized. Here we address this question in physiologically relevant striatal neurons and investigate how AC localization impacts downstream signaling function. We show that the major striatal AC isoforms are differentially sorted between ciliary and extraciliary domains of the plasma membrane, and that one isoform, AC9, is uniquely concentrated in endosomes. We identify key sorting determinants in the N-terminal cytoplasmic domain responsible for isoform-specific localization. We further show that AC9-containing endosomes accumulate activated dopamine receptors and form an elaborately intertwined network with juxtanuclear PKA stores bound to Golgi membranes. Finally, we provide evidence that endosomal localization enables AC9 to selectively elevate PKA activity in the nucleus relative to the cytoplasm. Together, these results reveal a precise spatial landscape of the cAMP cascade in neurons and a key role of AC localization in directing downstream PKA signaling to the nucleus.
- Published
- 2024
21. Profiling the proximal proteome of the activated μ-opioid receptor.
- Author
-
Polacco, Benjamin, Lobingier, Braden, Blythe, Emily, Abreu, Nohely, Khare, Prachi, Howard, Matthew, Gonzalez-Hernandez, Alberto, Xu, Jiewei, Li, Qiongyu, Novy, Brandon, Naing, Zun, Shoichet, Brian, Coyote-Maestas, Willow, Levitz, Joshua, Krogan, Nevan, Von Zastrow, Mark, and Hüttenhain, Ruth
- Subjects
Humans ,Endocytosis ,HEK293 Cells ,Proteome ,Proteomics ,Receptors ,G-Protein-Coupled ,Receptors ,Opioid ,mu ,Signal Transduction - Abstract
The μ-opioid receptor (μOR) represents an important target of therapeutic and abused drugs. So far, most understanding of μOR activity has focused on a subset of known signal transducers and regulatory molecules. Yet μOR signaling is coordinated by additional proteins in the interaction network of the activated receptor, which have largely remained invisible given the lack of technologies to interrogate these networks systematically. Here we describe a proteomics and computational approach to map the proximal proteome of the activated μOR and to extract subcellular location, trafficking and functional partners of G-protein-coupled receptor (GPCR) activity. We demonstrate that distinct opioid agonists exert differences in the μOR proximal proteome mediated by endocytosis and endosomal sorting. Moreover, we identify two new μOR network components, EYA4 and KCTD12, which are recruited on the basis of receptor-triggered G-protein activation and might form a previously unrecognized buffering system for G-protein activity broadly modulating cellular GPCR signaling.
- Published
- 2024
22. Unlocking opioid neuropeptide dynamics with genetically encoded biosensors
- Author
-
Dong, Chunyang, Gowrishankar, Raajaram, Jin, Yihan, He, Xinyi Jenny, Gupta, Achla, Wang, Huikun, Sayar-Atasoy, Nilüfer, Flores, Rodolfo J, Mahe, Karan, Tjahjono, Nikki, Liang, Ruqiang, Marley, Aaron, Or Mizuno, Grace, Lo, Darren K, Sun, Qingtao, Whistler, Jennifer L, Li, Bo, Gomes, Ivone, Von Zastrow, Mark, Tejeda, Hugo A, Atasoy, Deniz, Devi, Lakshmi A, Bruchas, Michael R, Banghart, Matthew R, and Tian, Lin
- Subjects
Pharmacology and Pharmaceutical Sciences ,Biomedical and Clinical Sciences ,Neurosciences ,Substance Misuse ,Pain Research ,Opioids ,Bioengineering ,Drug Abuse (NIDA only) ,1.1 Normal biological development and functioning ,Animals ,Biosensing Techniques ,Mice ,Optogenetics ,Neurons ,Humans ,Dynorphins ,Male ,Opioid Peptides ,HEK293 Cells ,Mice ,Inbred C57BL ,Brain ,Neuropeptides ,Receptors ,Opioid ,Electric Stimulation ,Reward ,Psychology ,Cognitive Sciences ,Neurology & Neurosurgery ,Biological psychology - Abstract
Neuropeptides are ubiquitous in the nervous system. Research into neuropeptides has been limited by a lack of experimental tools that allow for the precise dissection of their complex and diverse dynamics in a circuit-specific manner. Opioid peptides modulate pain, reward and aversion and as such have high clinical relevance. To illuminate the spatiotemporal dynamics of endogenous opioid signaling in the brain, we developed a class of genetically encoded fluorescence sensors based on kappa, delta and mu opioid receptors: κLight, δLight and µLight, respectively. We characterized the pharmacological profiles of these sensors in mammalian cells and in dissociated neurons. We used κLight to identify electrical stimulation parameters that trigger endogenous opioid release and the spatiotemporal scale of dynorphin volume transmission in brain slices. Using in vivo fiber photometry in mice, we demonstrated the utility of these sensors in detecting optogenetically driven opioid release and observed differential opioid release dynamics in response to fearful and rewarding conditions.
- Published
- 2024
23. Phospholipid scrambling induced by an ion channel/metabolite transporter complex.
- Author
-
Niu, Han, Maruoka, Masahiro, Noguchi, Yuki, Kosako, Hidetaka, and Suzuki, Jun
- Subjects
Humans ,Calcium ,Phospholipid Transfer Proteins ,Phospholipids ,HEK293 Cells ,Ion Channels ,Animals ,CRISPR-Cas Systems - Abstract
Cells establish the asymmetrical distribution of phospholipids and alter their distribution by phospholipid scrambling (PLS) to adapt to environmental changes. Here, we demonstrate that a protein complex, consisting of the ion channel Tmem63b and the thiamine transporter Slc19a2, induces PLS upon calcium (Ca2+) stimulation. Through revival screening using a CRISPR sgRNA library on high PLS cells, we identify Tmem63b as a PLS-inducing factor. Ca2+ stimulation-mediated PLS is suppressed by deletion of Tmem63b, while human disease-related Tmem63b mutants induce constitutive PLS. To search for a molecular link between Ca2+ stimulation and PLS, we perform revival screening on Tmem63b-overexpressing cells, and identify Slc19a2 and the Ca2+-activated K+ channel Kcnn4 as PLS-regulating factors. Deletion of either of these genes decreases PLS activity. Biochemical screening indicates that Tmem63b and Slc19a2 form a heterodimer. These results demonstrate that a Tmem63b/Slc19a2 heterodimer induces PLS upon Ca2+ stimulation, along with Kcnn4 activation.
- Published
- 2024
24. Subtype-specific conformational landscape of NMDA receptor gating.
- Author
-
Bleier, Julia, Furtado de Mendonca, Philipe, Habrian, Chris, Stanley, Cherise, Vyklicky, Vojtech, and Isacoff, Ehud
- Subjects
CP: Neuroscience ,FRET ,GluN1 ,GluN2 ,Grin1 ,Grin2 ,NMDA receptors ,allostery ,single-molecule FRET ,Receptors ,N-Methyl-D-Aspartate ,Humans ,Fluorescence Resonance Energy Transfer ,Animals ,Protein Conformation ,HEK293 Cells ,Ion Channel Gating ,Protein Subunits ,Protein Domains - Abstract
N-methyl-D-aspartate receptors are ionotropic glutamate receptors that mediate synaptic transmission and plasticity. Variable GluN2 subunits in diheterotetrameric receptors with identical GluN1 subunits set very different functional properties. To understand this diversity, we use single-molecule fluorescence resonance energy transfer (smFRET) to measure the conformations of the ligand binding domain and modulatory amino-terminal domain of the common GluN1 subunit in receptors with different GluN2 subunits. Our results demonstrate a strong influence of the GluN2 subunits on GluN1 rearrangements, both in non-agonized and partially agonized activation intermediates, which have been elusive to structural analysis, and in the fully liganded state. Chimeric analysis reveals structural determinants that contribute to these subtype differences. Our study provides a framework for understanding the conformational landscape that supports highly divergent levels of activity, desensitization, and agonist potency in receptors with different GluN2s and could open avenues for the development of subtype-specific modulators.
- Published
- 2024
25. Positive cooperativity in synergistic activation of Wnt proteins.
- Author
-
Bonnet, Clemence, Han, Christiana, Deng, Sophie, and Zheng, Jie
- Subjects
Cooperative binding ,Signalosome ,Wnt mimic ,Wnt/b-catenin signaling ,Wnt3a ,Humans ,HEK293 Cells ,Wnt Signaling Pathway ,Wnt3A Protein ,Wnt Proteins ,beta Catenin ,Ligands - Abstract
BACKGROUND: Wnt proteins are crucial for embryonic development, stem cell growth, and tissue regeneration. Wnt signaling pathway is activated when Wnt proteins bind to cell membrane receptors. METHODS AND RESULTS: We employed a luciferase reporter assay in HEK293STF cells to measure Wnt protein-induced signaling. We observed that Wnt3a uniquely promotes the Wnt/β-catenin pathway through positive cooperativity. Additionally, MFH-ND, a molecular mimic of Wnt ligands, markedly increased Wnt3a-induced signaling in a dose-responsive manner. This suggests that various Wnt ligands can synergistically enhance Wnt pathway activation. CONCLUSIONS: The study suggests the likelihood of various Wnt ligands coexisting in a single signalosome on the cell membrane, providing new insights into the complexities of Wnt signaling mechanisms.
- Published
- 2024
26. Degron-Based bioPROTACs for Controlling Signaling in CAR T Cells.
- Author
-
Kim, Matthew, Bhargava, Hersh, Shavey, Gavin, Lim, Wendell, El-Samad, Hana, and Ng, Andrew
- Subjects
CAR T cells ,mammalian synthetic biology ,targeted protein degradation ,Humans ,Degrons ,HEK293 Cells ,Proteasome Endopeptidase Complex ,Proteolysis ,Receptors ,Chimeric Antigen ,Signal Transduction ,T-Lymphocytes ,ZAP-70 Protein-Tyrosine Kinase - Abstract
Chimeric antigen receptor (CAR) T cells have made a tremendous impact in the clinic, but potent signaling through the CAR can be detrimental to treatment safety and efficacy. The use of protein degradation to control CAR signaling can address these issues in preclinical models. Existing strategies for regulating CAR stability rely on small molecules to induce systemic degradation. In contrast to small molecule regulation, genetic circuits offer a more precise method to control CAR signaling in an autonomous cell-by-cell fashion. Here, we describe a programmable protein degradation tool that adopts the framework of bioPROTACs, heterobifunctional proteins that are composed of a target recognition domain fused to a domain that recruits the endogenous ubiquitin proteasome system. We develop novel bioPROTACs that utilize a compact four-residue degron and demonstrate degradation of cytosolic and membrane protein targets using either a nanobody or synthetic leucine zipper as a protein binder. Our bioPROTACs exhibit potent degradation of CARs and can inhibit CAR signaling in primary human T cells. We demonstrate the utility of our bioPROTACs by constructing a genetic circuit to degrade the tyrosine kinase ZAP70 in response to recognition of a specific membrane-bound antigen. This circuit can disrupt CAR T cell signaling only in the presence of a specific cell population. These results suggest that bioPROTACs are powerful tools for expanding the CAR T cell engineering toolbox.
- Published
- 2024
27. Emissive Guanosine Analog Applicable for Real-Time Live Cell Imaging
- Author
-
Steinbuch, Kfir B, Cong, Deyuan, Rodriguez, Anthony J, and Tor, Yitzhak
- Subjects
Inorganic Chemistry ,Chemical Sciences ,Biological Sciences ,Humans ,DNA-Directed RNA Polymerases ,Viral Proteins ,Guanosine ,Microscopy ,Confocal ,Cell Survival ,HEK293 Cells ,Organic Chemistry ,Biological sciences ,Chemical sciences - Abstract
A new emissive guanosine analog CF3thG, constructed by a single trifluoromethylation step from the previously reported thG, displays red-shifted absorption and emission spectra compared to its precursor. The impact of solvent type and polarity on the photophysical properties of CF3thG suggests that the electronic effects of the trifluoromethyl group dominate its behavior and demonstrates its susceptibility to microenvironmental polarity changes. In vitro transcription initiations using T7 RNA polymerase, initiated with CF3thG, result in highly emissive 5'-labeled RNA transcripts, demonstrating the tolerance of the enzyme toward the analog. Viability assays with HEK293T cells displayed no detrimental effects at tested concentrations, indicating the safety of the analog for cellular applications. Live cell imaging of the free emissive guanosine analog using confocal microscopy was facilitated by its red-shifted absorption and emission and adequate brightness. Real-time live cell imaging demonstrated the release of the guanosine analog from HEK293T cells at concentration-gradient conditions, which was suppressed by the addition of guanosine.
- Published
- 2024
28. Systematic multi-trait AAV capsid engineering for efficient gene delivery.
- Author
-
Eid, Fatma-Elzahraa, Chen, Albert, Chan, Ken, Huang, Qin, Zheng, Qingxia, Tobey, Isabelle, Pacouret, Simon, Brauer, Pamela, Keyes, Casey, Powell, Megan, Johnston, Jencilin, Zhao, Binhui, Lage, Kasper, Tarantal, Alice, Chan, Yujia, and Deverman, Benjamin
- Subjects
Dependovirus ,Animals ,Humans ,Mice ,Genetic Vectors ,Capsid ,Capsid Proteins ,Liver ,Transduction ,Genetic ,Gene Transfer Techniques ,Machine Learning ,Genetic Therapy ,Macaca ,Hepatocytes ,HEK293 Cells ,Genetic Engineering - Abstract
Broadening gene therapy applications requires manufacturable vectors that efficiently transduce target cells in humans and preclinical models. Conventional selections of adeno-associated virus (AAV) capsid libraries are inefficient at searching the vast sequence space for the small fraction of vectors possessing multiple traits essential for clinical translation. Here, we present Fit4Function, a generalizable machine learning (ML) approach for systematically engineering multi-trait AAV capsids. By leveraging a capsid library that uniformly samples the manufacturable sequence space, reproducible screening data are generated to train accurate sequence-to-function models. Combining six models, we designed a multi-trait (liver-targeted, manufacturable) capsid library and validated 88% of library variants on all six predetermined criteria. Furthermore, the models, trained only on mouse in vivo and human in vitro Fit4Function data, accurately predicted AAV capsid variant biodistribution in macaque. Top candidates exhibited production yields comparable to AAV9, efficient murine liver transduction, up to 1000-fold greater human hepatocyte transduction, and increased enrichment relative to AAV9 in a screen for liver transduction in macaques. The Fit4Function strategy ultimately makes it possible to predict cross-species traits of peptide-modified AAV capsids and is a critical step toward assembling an ML atlas that predicts AAV capsid performance across dozens of traits.
- Published
- 2024
29. A proximity proteomics pipeline with improved reproducibility and throughput.
- Author
-
Zhong, Xiaofang, Li, Qiongyu, Polacco, Benjamin, Patil, Trupti, Marley, Aaron, Foussard, Helene, Khare, Prachi, Vartak, Rasika, Xu, Jiewei, DiBerto, Jeffrey, Roth, Bryan, Eckhardt, Manon, von Zastrow, Mark, Krogan, Nevan, and Hüttenhain, Ruth
- Subjects
APEX2-based Proximity Labeling ,G Protein-Coupled Receptor ,Protein–Protein Interaction ,Proximity Proteomics ,Subcellular Proteomics ,Proteomics ,Biotinylation ,Reproducibility of Results ,Humans ,Proteome ,Mass Spectrometry ,HEK293 Cells - Abstract
Proximity labeling (PL) via biotinylation coupled with mass spectrometry (MS) captures spatial proteomes in cells. Large-scale processing requires a workflow minimizing hands-on time and enhancing quantitative reproducibility. We introduced a scalable PL pipeline integrating automated enrichment of biotinylated proteins in a 96-well plate format. Combining this with optimized quantitative MS based on data-independent acquisition (DIA), we increased sample throughput and improved protein identification and quantification reproducibility. We applied this pipeline to delineate subcellular proteomes across various compartments. Using the 5HT2A serotonin receptor as a model, we studied temporal changes of proximal interaction networks induced by receptor activation. In addition, we modified the pipeline for reduced sample input to accommodate CRISPR-based gene knockout, assessing dynamics of the 5HT2A network in response to perturbation of selected interactors. This PL approach is universally applicable to PL proteomics using biotinylation-based PL enzymes, enhancing throughput and reproducibility of standard protocols.
- Published
- 2024
30. Factors affecting protein recovery during Hsp40 affinity profiling.
- Author
-
Montoya, Maureen, Quanrud, Guy, Mei, Liangyong, Moñtano, José, Hong, Caleb, and Genereux, Joseph
- Subjects
AP-MS ,DNAJB1 ,DNAJB8 ,Hsp40 ,Misfolded proteins ,Proteomics ,HSP40 Heat-Shock Proteins ,Humans ,HEK293 Cells ,Proteomics ,Protein Binding ,HSP70 Heat-Shock Proteins ,Protein Stability ,Protein Folding - Abstract
The identification and quantification of misfolded proteins from complex mixtures is important for biological characterization and disease diagnosis, but remains a major bioanalytical challenge. We have developed Hsp40 Affinity Profiling as a bioanalytical approach to profile protein stability in response to cellular stress. In this assay, we ectopically introduce the Hsp40 FlagDNAJB8H31Q into cells and use quantitative proteomics to determine how protein affinity for DNAJB8 changes in the presence of cellular stress, without regard for native clients. Herein, we evaluate potential approaches to improve the performance of this bioanalytical assay. We find that although intracellular crosslinking increases recovery of protein interactors, this is not enough to overcome the relative drop in DNAJB8 recovery. While the J-domain promotes Hsp70 association, it does not affect the yield of protein association with DNAJB8 under basal conditions. By contrast, crosslinking and J-domain ablation both substantially increase relative protein interactor recovery with the structurally distinct Class B Hsp40 DNAJB1 but are completely compensated by poorer yield of DNAJB1 itself. Cellular thermal stress promotes increased affinity between DNAJB8H31Q and interacting proteins, as expected for interactions driven by recognition of misfolded proteins. DNAJB8WT does not demonstrate such a property, suggesting that under stress misfolded proteins are handed off to Hsp70. Hence, we find that DNAJB8H31Q is still our most effective recognition element for the recovery of destabilized client proteins following cellular stress.
- Published
- 2024
31. Cardiolipin remodeling maintains the inner mitochondrial membrane in cells with saturated lipidomes.
- Author
-
Venkatraman, Kailash and Budin, Itay
- Subjects
Barth syndrome ,cardiolipin ,lipid saturation ,mitochondria ,phospholipids ,Cardiolipins ,Humans ,Saccharomyces cerevisiae ,Mitochondrial Membranes ,HEK293 Cells ,Saccharomyces cerevisiae Proteins ,Lipidomics ,Fatty Acids ,Barth Syndrome ,Acyltransferases ,Phospholipases - Abstract
Cardiolipin (CL) is a unique, four-chain phospholipid synthesized in the inner mitochondrial membrane (IMM). The acyl chain composition of CL is regulated through a remodeling pathway, whose loss causes mitochondrial dysfunction in Barth syndrome (BTHS). Yeast has been used extensively as a model system to characterize CL metabolism, but mutants lacking its two remodeling enzymes, Cld1p and Taz1p, exhibit mild structural and respiratory phenotypes compared to mammalian cells. Here, we show an essential role for CL remodeling in the structure and function of the IMM in yeast grown under reduced oxygenation. Microaerobic fermentation, which mimics natural yeast environments, caused the accumulation of saturated fatty acids and, under these conditions, remodeling mutants showed a loss of IMM ultrastructure. We extended this observation to HEK293 cells, where phospholipase A2 inhibition by Bromoenol lactone resulted in respiratory dysfunction and cristae loss upon mild treatment with exogenous saturated fatty acids. In microaerobic yeast, remodeling mutants accumulated unremodeled, saturated CL, but also displayed reduced total CL levels, highlighting the interplay between saturation and CL biosynthesis and/or breakdown. We identified the mitochondrial phospholipase A1 Ddl1p as a regulator of CL levels, and those of its precursors phosphatidylglycerol and phosphatidic acid, under these conditions. Loss of Ddl1p partially rescued IMM structure in cells unable to initiate CL remodeling and had differing lipidomic effects depending on oxygenation. These results introduce a revised yeast model for investigating CL remodeling and suggest that its structural functions are dependent on the overall lipid environment in the mitochondrion.
- Published
- 2024
32. A novel reporter for helicase activity in translation uncovers DDX3X interactions
- Author
-
Wilkins, Kevin, Schroeder, Till, Gu, Sohyun, Revalde, Jezrael Lafuente, and Floor, Stephen N
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,1.1 Normal biological development and functioning ,Generic health relevance ,DEAD-box RNA Helicases ,Humans ,5' Untranslated Regions ,Protein Biosynthesis ,Genes ,Reporter ,Nucleic Acid Conformation ,RNA ,Messenger ,HEK293 Cells ,Protein Binding ,RNA helicases ,RNA structure ,reporter genes ,translational control ,Developmental Biology ,Biochemistry and cell biology - Abstract
DDX3X regulates the translation of a subset of human transcripts containing complex 5' untranslated regions (5' UTRs). In this study, we developed the helicase activity reporter for translation (HART), which uses DDX3X-sensitive 5' UTRs to measure DDX3X-mediated translational activity in cells. To directly measure RNA structure in DDX3X-dependent mRNAs, we used SHAPE-MaP to determine the secondary structures present in DDX3X-sensitive 5' UTRs and then used HART to investigate how sequence alterations influence DDX3X sensitivity. Additionally, we identified residues 38-44 as potential mediators of DDX3X's interaction with the translational machinery. HART revealed that both DDX3X's association with the translational machinery and its helicase activity are required for its function in promoting the translation of DDX3X-sensitive 5' UTRs. These findings suggest DDX3X plays a crucial role in regulating translation through its interaction with the translational machinery during ribosome scanning and establish the HART reporter as a robust, lentivirally encoded, colorimetric measurement of DDX3X-dependent translation in cells.
- Published
- 2024
33. Cilia-enriched oxysterol 7β,27-DHC is required for polycystin ion channel activation.
- Author
-
Ha, Kodaji, Mundt-Machado, Nadine, Bisignano, Paola, Pinedo, Aide, Raleigh, David, Loeb, Gabriel, Reiter, Jeremy, Cao, Erhu, and Delling, Markus
- Subjects
Cilia ,TRPP Cation Channels ,Animals ,Humans ,Polycystic Kidney ,Autosomal Dominant ,Oxysterols ,Patch-Clamp Techniques ,HEK293 Cells ,Mutation ,Kidney ,Mice ,Binding Sites - Abstract
Polycystin-1 (PC-1) and PC-2 form a heteromeric ion channel complex that is abundantly expressed in primary cilia of renal epithelial cells. This complex functions as a non-selective cation channel, and mutations within the polycystin complex cause autosomal dominant polycystic kidney disease (ADPKD). The spatial and temporal regulation of the polycystin complex within the ciliary membrane remains poorly understood. Using both whole-cell and ciliary patch-clamp recordings, we identify a cilia-enriched oxysterol, 7β,27-dihydroxycholesterol (DHC), that serves as a necessary activator of the polycystin complex. We further identify an oxysterol-binding pocket within PC-2 and showed that mutations within this binding pocket disrupt 7β,27-DHC-dependent polycystin activation. Pharmacologic and genetic inhibition of oxysterol synthesis reduces channel activity in primary cilia. In summary, our findings reveal a regulator of the polycystin complex. This oxysterol-binding pocket in PC-2 may provide a specific target for potential ADPKD therapeutics.
- Published
- 2024
34. Structural basis for the H2AK119ub1-specific DNMT3A-nucleosome interaction.
- Author
-
Chen, Xinyi, Guo, Yiran, Zhao, Ting, Lu, Jiuwei, Fang, Jian, Wang, Yinsheng, Wang, Gang, and Song, Jikui
- Subjects
Nucleosomes ,DNA Methyltransferase 3A ,DNA (Cytosine-5-)-Methyltransferases ,Histones ,Humans ,DNA Methylation ,Protein Binding ,Cryoelectron Microscopy ,Animals ,Mice ,Ubiquitination ,Polycomb Repressive Complex 2 ,HEK293 Cells ,Models ,Molecular - Abstract
Isoform 1 of DNA methyltransferase DNMT3A (DNMT3A1) specifically recognizes nucleosome monoubiquitylated at histone H2A lysine-119 (H2AK119ub1) for establishment of DNA methylation. Mis-regulation of this process may cause aberrant DNA methylation and pathogenesis. However, the molecular basis underlying DNMT3A1-nucleosome interaction remains elusive. Here we report the cryo-EM structure of DNMT3A1s ubiquitin-dependent recruitment (UDR) fragment complexed with H2AK119ub1-modified nucleosome. DNMT3A1 UDR occupies an extensive nucleosome surface, involving the H2A-H2B acidic patch, a surface groove formed by H2A and H3, nucleosomal DNA, and H2AK119ub1. The DNMT3A1 UDRs interaction with H2AK119ub1 affects the functionality of DNMT3A1 in cells in a context-dependent manner. Our structural and biochemical analysis also reveals competition between DNMT3A1 and JARID2, a cofactor of polycomb repression complex 2 (PRC2), for nucleosome binding, suggesting the interplay between different epigenetic pathways. Together, this study reports a molecular basis for H2AK119ub1-dependent DNMT3A1-nucleosome association, with important implications in DNMT3A1-mediated DNA methylation in development.
- Published
- 2024
35. Engineering programmable material-to-cell pathways via synthetic notch receptors to spatially control differentiation in multicellular constructs.
- Author
-
Garibyan, Mher, Hoffman, Tyler, Makaske, Thijs, Do, Stephanie, Wu, Yifan, Williams, Brian, March, Alexander, Cho, Nathan, Pedroncelli, Nicolas, Lima, Ricardo, Soto, Jennifer, Jackson, Brooke, Santoso, Jeffrey, Khademhosseini, Ali, Thomson, Matt, Li, Song, McCain, Megan, and Morsut, Leonardo
- Subjects
Receptors ,Notch ,Cell Differentiation ,Tissue Engineering ,Animals ,Humans ,Signal Transduction ,Mice ,Extracellular Matrix ,Fibroblasts ,Extracellular Matrix Proteins ,Ligands ,Tissue Scaffolds ,Muscle ,Skeletal ,Endothelial Cells ,HEK293 Cells - Abstract
Synthetic Notch (synNotch) receptors are genetically encoded, modular synthetic receptors that enable mammalian cells to detect environmental signals and respond by activating user-prescribed transcriptional programs. Although some materials have been modified to present synNotch ligands with coarse spatial control, applications in tissue engineering generally require extracellular matrix (ECM)-derived scaffolds and/or finer spatial positioning of multiple ligands. Thus, we develop here a suite of materials that activate synNotch receptors for generalizable engineering of material-to-cell signaling. We genetically and chemically fuse functional synNotch ligands to ECM proteins and ECM-derived materials. We also generate tissues with microscale precision over four distinct reporter phenotypes by culturing cells with two orthogonal synNotch programs on surfaces microcontact-printed with two synNotch ligands. Finally, we showcase applications in tissue engineering by co-transdifferentiating fibroblasts into skeletal muscle or endothelial cell precursors in user-defined micropatterns. These technologies provide avenues for spatially controlling cellular phenotypes in mammalian tissues.
- Published
- 2024
36. The Chlamydia trachomatis Inc Tri1 interacts with TRAF7 to displace native TRAF7 interacting partners.
- Author
-
Herrera, Clara, McMahon, Eleanor, Swaney, Danielle, Sherry, Jessica, Pha, Khavong, Adams-Boone, Kathleen, Johnson, Jeffrey, Krogan, Nevan, Stevers, Meredith, Solomon, David, Elwell, Cherilyn, and Engel, Joanne
- Subjects
Chlamydia trachomatis ,MEKK2 ,MEKK3 ,TRAF7 ,WD40 ,host-pathogen interaction ,inclusion membrane protein ,mass spectrometry ,Humans ,Chlamydia trachomatis ,Host-Pathogen Interactions ,HeLa Cells ,Bacterial Proteins ,Chlamydia Infections ,Signal Transduction ,Tumor Necrosis Factor Receptor-Associated Peptides and Proteins ,Immunity ,Innate ,Protein Binding ,Membrane Proteins ,HEK293 Cells - Abstract
Chlamydia trachomatis is the leading cause of bacterial sexually transmitted infections in the USA and of preventable blindness worldwide. This obligate intracellular pathogen replicates within a membrane-bound inclusion, but how it acquires nutrients from the host while avoiding detection by the innate immune system is incompletely understood. C. trachomatis accomplishes this in part through the translocation of a unique set of effectors into the inclusion membrane, the inclusion membrane proteins (Incs). Incs are ideally positioned at the host-pathogen interface to reprogram host signaling by redirecting proteins or organelles to the inclusion. Using a combination of co-affinity purification, immunofluorescence confocal imaging, and proteomics, we characterize the interaction between an early-expressed Inc of unknown function, Tri1, and tumor necrosis factor receptor-associated factor 7 (TRAF7). TRAF7 is a multi-domain protein with a RING finger ubiquitin ligase domain and a C-terminal WD40 domain. TRAF7 regulates several innate immune signaling pathways associated with C. trachomatis infection and is mutated in a subset of tumors. We demonstrate that Tri1 and TRAF7 specifically interact during infection and that TRAF7 is recruited to the inclusion. We further show that the predicted coiled-coil domain of Tri1 is necessary to interact with the TRAF7 WD40 domain. Finally, we demonstrate that Tri1 displaces the native TRAF7 binding partners, mitogen-activated protein kinase kinase kinase 2 (MEKK2), and MEKK3. Together, our results suggest that by displacing TRAF7 native binding partners, Tri1 has the capacity to alter TRAF7 signaling during C. trachomatis infection.IMPORTANCEChlamydia trachomatis is the leading cause of bacterial sexually transmitted infections in the USA and preventable blindness worldwide. Although easily treated with antibiotics, the vast majority of infections are asymptomatic and therefore go untreated, leading to infertility and blindness. This obligate intracellular pathogen evades the immune response, which contributes to these outcomes. Here, we characterize the interaction between a C. trachomatis-secreted effector, Tri1, and a host protein involved in innate immune signaling, TRAF7. We identified host proteins that bind to TRAF7 and demonstrated that Tri1 can displace these proteins upon binding to TRAF7. Remarkably, the region of TRAF7 to which these host proteins bind is often mutated in a subset of human tumors. Our work suggests a mechanism by which Tri1 may alter TRAF7 signaling and has implications not only in the pathogenesis of C. trachomatis infections but also in understanding the role of TRAF7 in cancer.
- Published
- 2024
37. A designed ankyrin-repeat protein that targets Parkinsons disease-associated LRRK2.
- Author
-
Dederer, Verena, Sanz Murillo, Marta, Karasmanis, Eva, Hatch, Kathryn, Chatterjee, Deep, Preuss, Franziska, Abdul Azeez, Kamal, Nguyen, Landon, Galicia, Christian, Dreier, Birgit, Plückthun, Andreas, Versees, Wim, Mathea, Sebastian, Leschziner, Andres, Reck-Peterson, Samara, and Knapp, Stefan
- Subjects
DARPin ,LRRK2 ,Parkinson’s disease ,Rab8a ,WD40 ,cryo-electron microscopy ,kinase ,kinase inhibitor ,microtubule ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,Humans ,Ankyrin Repeat ,Parkinson Disease ,HEK293 Cells ,rab GTP-Binding Proteins ,Phosphorylation ,Cryoelectron Microscopy ,Protein Binding - Abstract
Leucine rich repeat kinase 2 (LRRK2) is a large multidomain protein containing two catalytic domains, a kinase and a GTPase, as well as protein interactions domains, including a WD40 domain. The association of increased LRRK2 kinase activity with both the familial and sporadic forms of Parkinsons disease has led to an intense interest in determining its cellular function. However, small molecule probes that can bind to LRRK2 and report on or affect its cellular activity are needed. Here, we report the identification and characterization of the first high-affinity LRRK2-binding designed ankyrin-repeat protein (DARPin), named E11. Using cryo-EM, we show that DARPin E11 binds to the LRRK2 WD40 domain. LRRK2 bound to DARPin E11 showed improved behavior on cryo-EM grids, resulting in higher resolution LRRK2 structures. DARPin E11 did not affect the catalytic activity of a truncated form of LRRK2 in vitro but decreased the phosphorylation of Rab8A, a LRRK2 substrate, in cells. We also found that DARPin E11 disrupts the formation of microtubule-associated LRRK2 filaments in cells, which are known to require WD40-based dimerization. Thus, DARPin E11 is a new tool to explore the function and dysfunction of LRRK2 and guide the development of LRRK2 kinase inhibitors that target the WD40 domain instead of the kinase.
- Published
- 2024
38. Ribosome rescue factor PELOTA modulates translation start site choice for C/EBPα protein isoforms.
- Author
-
Fernandez, Samantha, Ferguson, Lucas, and Ingolia, Nicholas
- Subjects
Protein Isoforms ,Humans ,Ribosomes ,CCAAT-Enhancer-Binding Protein-alpha ,Protein Biosynthesis ,RNA ,Messenger ,Animals ,Peptide Chain Initiation ,Translational ,Mice ,TOR Serine-Threonine Kinases ,HEK293 Cells - Abstract
Translation initiation at alternative start sites can dynamically control the synthesis of two or more functionally distinct protein isoforms from a single mRNA. Alternate isoforms of the developmental transcription factor CCAAT/enhancer-binding protein α (C/EBPα) produced from different start sites exert opposing effects during myeloid cell development. This choice between alternative start sites depends on sequence features of the CEBPA transcript, including a regulatory uORF, but the molecular basis is not fully understood. Here, we identify the factors that affect C/EBPα isoform choice using a sensitive and quantitative two-color fluorescent reporter coupled with CRISPRi screening. Our screen uncovered a role of the ribosome rescue factor PELOTA (PELO) in promoting the expression of the longer C/EBPα isoform by directly removing inhibitory unrecycled ribosomes and through indirect effects mediated by the mechanistic target of rapamycin kinase. Our work uncovers further links between ribosome recycling and translation reinitiation that regulate a key transcription factor, with implications for normal hematopoiesis and leukemogenesis.
- Published
- 2024
39. A RAB7A phosphoswitch coordinates Rubicon Homology protein regulation of Parkin-dependent mitophagy.
- Author
-
Tudorica, Dan, Basak, Bishal, Puerta Cordova, Alexia, Khuu, Grace, Rose, Kevin, Lazarou, Michael, Holzbaur, Erika, and Hurley, James
- Subjects
Mitophagy ,Humans ,rab7 GTP-Binding Proteins ,Phosphorylation ,Ubiquitin-Protein Ligases ,Protein Serine-Threonine Kinases ,rab GTP-Binding Proteins ,HeLa Cells ,Protein Binding ,Intracellular Signaling Peptides and Proteins ,Autophagy-Related Proteins ,Mitochondria ,HEK293 Cells - Abstract
Activation of PINK1 and Parkin in response to mitochondrial damage initiates a response that includes phosphorylation of RAB7A at Ser72. Rubicon is a RAB7A binding negative regulator of autophagy. The structure of the Rubicon:RAB7A complex suggests that phosphorylation of RAB7A at Ser72 would block Rubicon binding. Indeed, in vitro phosphorylation of RAB7A by TBK1 abrogates Rubicon:RAB7A binding. Pacer, a positive regulator of autophagy, has an RH domain with a basic triad predicted to bind an introduced phosphate. Consistent with this, Pacer-RH binds to phosho-RAB7A but not to unphosphorylated RAB7A. In cells, mitochondrial depolarization reduces Rubicon:RAB7A colocalization whilst recruiting Pacer to phospho-RAB7A-positive puncta. Pacer knockout reduces Parkin mitophagy with little effect on bulk autophagy or Parkin-independent mitophagy. Rescue of Parkin-dependent mitophagy requires the intact pRAB7A phosphate-binding basic triad of Pacer. Together these structural and functional data support a model in which the TBK1-dependent phosphorylation of RAB7A serves as a switch, promoting mitophagy by relieving Rubicon inhibition and favoring Pacer activation.
- Published
- 2024
40. HIV-1 Vpr combats the PU.1-driven antiviral response in primary human macrophages.
- Author
-
Virgilio, Maria, Ramnani, Barkha, Chen, Thomas, Disbennett, W, Lubow, Jay, Welch, Joshua, and Collins, Kathleen
- Subjects
Humans ,Macrophages ,vpr Gene Products ,Human Immunodeficiency Virus ,HIV-1 ,Trans-Activators ,Proto-Oncogene Proteins ,Immunity ,Innate ,Ubiquitin-Protein Ligases ,HIV Infections ,HEK293 Cells ,Virion ,Protein Serine-Threonine Kinases - Abstract
HIV-1 Vpr promotes efficient spread of HIV-1 from macrophages to T cells by transcriptionally downmodulating restriction factors that target HIV-1 Envelope protein (Env). Here we find that Vpr induces broad transcriptomic changes by targeting PU.1, a transcription factor necessary for expression of host innate immune response genes, including those that target Env. Consistent with this, we find silencing PU.1 in infected macrophages lacking Vpr rescues Env. Vpr downmodulates PU.1 through a proteasomal degradation pathway that depends on physical interactions with PU.1 and DCAF1, a component of the Cul4A E3 ubiquitin ligase. The capacity for Vpr to target PU.1 is highly conserved across primate lentiviruses. In addition to impacting infected cells, we find that Vpr suppresses expression of innate immune response genes in uninfected bystander cells, and that virion-associated Vpr can degrade PU.1. Together, we demonstrate Vpr counteracts PU.1 in macrophages to blunt antiviral immune responses and promote viral spread.
- Published
- 2024
41. Structural insights into GABAA receptor potentiation by Quaalude.
- Author
-
Chojnacka, Weronika, Teng, Jinfeng, Kim, Jeong, Jensen, Anders, and Hibbs, Ryan
- Subjects
Receptors ,GABA-A ,Binding Sites ,Cryoelectron Microscopy ,Humans ,Animals ,Etomidate ,Propofol ,Quinazolinones ,Allosteric Regulation ,HEK293 Cells ,Hypnotics and Sedatives - Abstract
Methaqualone, a quinazolinone marketed commercially as Quaalude, is a central nervous system depressant that was used clinically as a sedative-hypnotic, then became a notorious recreational drug in the 1960s-80s. Due to its high abuse potential, medical use of methaqualone was eventually prohibited, yet it persists as a globally abused substance. Methaqualone principally targets GABAA receptors, which are the major inhibitory neurotransmitter-gated ion channels in the brain. The restricted status and limited accessibility of methaqualone have contributed to its pharmacology being understudied. Here, we use cryo-EM to localize the GABAA receptor binding sites of methaqualone and its more potent derivative, PPTQ, to the same intersubunit transmembrane sites targeted by the general anesthetics propofol and etomidate. Both methaqualone and PPTQ insert more deeply into subunit interfaces than the previously-characterized modulators. Binding of quinazolinones to this site results in widening of the extracellular half of the ion-conducting pore, following a trend among positive allosteric modulators in destabilizing the hydrophobic activation gate in the pore as a mechanism for receptor potentiation. These insights shed light on the underexplored pharmacology of quinazolinones and further elucidate the molecular mechanisms of allosteric GABAA receptor modulation through transmembrane binding sites.
- Published
- 2024
42. Haploinsufficiency underlies the neurodevelopmental consequences of SLC6A1 variants.
- Author
-
Silva, Dina, Trinidad, Marena, Ljungdahl, Alicia, Revalde, Jezrael, Berguig, Geoffrey, Wallace, William, Patrick, Cory, Bomba, Lorenzo, Arkin, Michelle, Dong, Shan, Estrada, Karol, Hutchinson, Keino, LeBowitz, Jonathan, Schlessinger, Avner, Johannesen, Katrine, Møller, Rikke, Giacomini, Kathleen, Froelich, Steven, Sanders, Stephan, and Wuster, Arthur
- Subjects
GABA uptake ,GAT-1 ,GAT1 ,SLC6A1 ,autism spectrum disorders ,epilepsy with myoclonic-atonic seizures ,missense vulnerability ,neurodevelopmental delay ,Humans ,GABA Plasma Membrane Transport Proteins ,Haploinsufficiency ,Mutation ,Missense ,gamma-Aminobutyric Acid ,Neurodevelopmental Disorders ,Developmental Disabilities ,Autistic Disorder ,HEK293 Cells - Abstract
Heterozygous variants in SLC6A1, encoding the GAT-1 GABA transporter, are associated with seizures, developmental delay, and autism. The majority of affected individuals carry missense variants, many of which are recurrent germline de novo mutations, raising the possibility of gain-of-function or dominant-negative effects. To understand the functional consequences, we performed an in vitro GABA uptake assay for 213 unique variants, including 24 control variants. De novo variants consistently resulted in a decrease in GABA uptake, in keeping with haploinsufficiency underlying all neurodevelopmental phenotypes. Where present, ClinVar pathogenicity reports correlated well with GABA uptake data; the functional data can inform future reports for the remaining 72% of unscored variants. Surface localization was assessed for 86 variants; two-thirds of loss-of-function missense variants prevented GAT-1 from being present on the membrane while GAT-1 was on the surface but with reduced activity for the remaining third. Surprisingly, recurrent de novo missense variants showed moderate loss-of-function effects that reduced GABA uptake with no evidence for dominant-negative or gain-of-function effects. Using linear regression across multiple missense severity scores to extrapolate the functional data to all potential SLC6A1 missense variants, we observe an abundance of GAT-1 residues that are sensitive to substitution. The extent of this missense vulnerability accounts for the clinically observed missense enrichment; overlap with hypermutable CpG sites accounts for the recurrent missense variants. Strategies to increase the expression of the wild-type SLC6A1 allele are likely to be beneficial across neurodevelopmental disorders, though the developmental stage and extent of required rescue remain unknown.
- Published
- 2024
43. Growth factor–dependent phosphorylation of Gαi shapes canonical signaling by G protein–coupled receptors
- Author
-
Roy, Suchismita, Sinha, Saptarshi, Silas, Ananta James, Ghassemian, Majid, Kufareva, Irina, and Ghosh, Pradipta
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,1.1 Normal biological development and functioning ,Generic health relevance ,Phosphorylation ,Signal Transduction ,Humans ,HEK293 Cells ,GTP-Binding Protein alpha Subunits ,Gi-Go ,Receptors ,CXCR4 ,Epidermal Growth Factor ,Receptors ,G-Protein-Coupled ,Animals ,Biochemistry and cell biology - Abstract
A long-standing question in the field of signal transduction is how distinct signaling pathways interact with each other to control cell behavior. Growth factor receptors and G protein-coupled receptors (GPCRs) are the two major signaling hubs in eukaryotes. Given that the mechanisms by which they signal independently have been extensively characterized, we investigated how they may cross-talk with each other. Using linear ion trap mass spectrometry and cell-based biophysical, biochemical, and phenotypic assays, we found at least three distinct ways in which epidermal growth factor affected canonical G protein signaling by the Gi-coupled GPCR CXCR4 through the phosphorylation of Gαi. Phosphomimicking mutations in two residues in the αE helix of Gαi (tyrosine-154/tyrosine-155) suppressed agonist-induced Gαi activation while promoting constitutive Gβγ signaling. Phosphomimicking mutations in the P loop (serine-44, serine-47, and threonine-48) suppressed Gi activation entirely, thus completely segregating growth factor and GPCR pathways. As expected, most of the phosphorylation events appeared to affect intrinsic properties of Gαi proteins, including conformational stability, nucleotide binding, and the ability to associate with and to release Gβγ. However, one phosphomimicking mutation, targeting the carboxyl-terminal residue tyrosine-320, promoted mislocalization of Gαi from the plasma membrane, a previously uncharacterized mechanism of suppressing GPCR signaling through G protein subcellular compartmentalization. Together, these findings elucidate not only how growth factor and chemokine signals cross-talk through the phosphorylation-dependent modulation of Gαi but also how such cross-talk may generate signal diversity.
- Published
- 2024
44. Rapid DNA unwinding accelerates genome editing by engineered CRISPR-Cas9
- Author
-
Eggers, Amy R, Chen, Kai, Soczek, Katarzyna M, Tuck, Owen T, Doherty, Erin E, Xu, Bryant, Trinidad, Marena I, Thornton, Brittney W, Yoon, Peter H, and Doudna, Jennifer A
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Gene Therapy ,Biotechnology ,Genetics ,2.1 Biological and endogenous factors ,1.1 Normal biological development and functioning ,Cancer ,Generic health relevance ,Humans ,Bacterial Proteins ,CRISPR-Associated Protein 9 ,CRISPR-Cas Systems ,Cryoelectron Microscopy ,DNA ,Gene Editing ,Geobacillus stearothermophilus ,HEK293 Cells ,Protein Domains ,Genome ,Human ,Models ,Molecular ,Protein Structure ,Tertiary ,Nucleic Acid Conformation ,Biocatalysis ,Magnesium ,CRISPR-Cas ,Cas9 engineering ,DNA unwinding ,GeoCas9 ,R-loop formation ,WED domain ,cryo-EM ,genome editing ,iGeoCas9 ,magnesium ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
Thermostable clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated (Cas9) enzymes could improve genome-editing efficiency and delivery due to extended protein lifetimes. However, initial experimentation demonstrated Geobacillus stearothermophilus Cas9 (GeoCas9) to be virtually inactive when used in cultured human cells. Laboratory-evolved variants of GeoCas9 overcome this natural limitation by acquiring mutations in the wedge (WED) domain that produce >100-fold-higher genome-editing levels. Cryoelectron microscopy (cryo-EM) structures of the wild-type and improved GeoCas9 (iGeoCas9) enzymes reveal extended contacts between the WED domain of iGeoCas9 and DNA substrates. Biochemical analysis shows that iGeoCas9 accelerates DNA unwinding to capture substrates under the magnesium-restricted conditions typical of mammalian but not bacterial cells. These findings enabled rational engineering of other Cas9 orthologs to enhance genome-editing levels, pointing to a general strategy for editing enzyme improvement. Together, these results uncover a new role for the Cas9 WED domain in DNA unwinding and demonstrate how accelerated target unwinding dramatically improves Cas9-induced genome-editing activity.
- Published
- 2024
45. De novo-designed transmembrane proteins bind and regulate a cytokine receptor.
- Author
-
Mravic, Marco, He, Li, Kratochvil, Huong, Hu, Hailin, Nick, Sarah, Bai, Weiya, Edwards, Anne, DiMaio, Daniel, Degrado, William, Jo, Hyunil, and Wu, Yibing
- Subjects
Humans ,Membrane Proteins ,Receptors ,Erythropoietin ,Protein Binding ,Models ,Molecular ,Cell Proliferation ,Receptors ,Cytokine ,Amino Acid Sequence ,Protein Multimerization ,Animals ,HEK293 Cells - Abstract
Transmembrane (TM) domains as simple as a single span can perform complex biological functions using entirely lipid-embedded chemical features. Computational design has the potential to generate custom tool molecules directly targeting membrane proteins at their functional TM regions. Thus far, designed TM domain-targeting agents have been limited to mimicking the binding modes and motifs of natural TM interaction partners. Here, we demonstrate the design of de novo TM proteins targeting the erythropoietin receptor (EpoR) TM domain in a custom binding topology competitive with receptor homodimerization. The TM proteins expressed in mammalian cells complex with EpoR and inhibit erythropoietin-induced cell proliferation. In vitro, the synthetic TM domain complex outcompetes EpoR homodimerization. Structural characterization reveals that the complex involves the intended amino acids and agrees with our designed molecular model of antiparallel TM helices at 1:1 stoichiometry. Thus, membrane protein TM regions can now be targeted in custom-designed topologies.
- Published
- 2024
46. Illuminating the function of the orphan transporter, SLC22A10, in humans and other primates.
- Author
-
Ferrández-Peral, Luis, Alentorn-Moron, Pol, Fontsere, Claudia, Ceylan, Merve, Koleske, Megan, Handin, Niklas, Artegoitia, Virginia, Lara, Giovanni, Chien, Huan-Chieh, Zhou, Xujia, Dainat, Jacques, Zalevsky, Arthur, Sali, Andrej, Brand, Colin, Wolfreys, Finn, Yang, Jia, Capra, John, Artursson, Per, Marquès-Bonet, Tomàs, Gestwicki, Jason, Giacomini, Kathleen, Newman, John, and Yee, Sook Wah
- Subjects
Animals ,Humans ,Amino Acid Sequence ,Estradiol ,HEK293 Cells ,Hominidae ,Mutation ,Missense ,Organic Cation Transport Proteins ,Primates ,Pseudogenes ,Substrate Specificity - Abstract
SLC22A10 is an orphan transporter with unknown substrates and function. The goal of this study is to elucidate its substrate specificity and functional characteristics. In contrast to orthologs from great apes, human SLC22A10, tagged with green fluorescent protein, is not expressed on the plasma membrane. Cells expressing great ape SLC22A10 orthologs exhibit significant accumulation of estradiol-17β-glucuronide, unlike those expressing human SLC22A10. Sequence alignments reveal a proline at position 220 in humans, which is a leucine in great apes. Replacing proline with leucine in SLC22A10-P220L restores plasma membrane localization and uptake function. Neanderthal and Denisovan genomes show proline at position 220, akin to modern humans, indicating functional loss during hominin evolution. Human SLC22A10 is a unitary pseudogene due to a fixed missense mutation, P220, while in great apes, its orthologs transport sex steroid conjugates. Characterizing SLC22A10 across species sheds light on its biological role, influencing organism development and steroid homeostasis.
- Published
- 2024
47. A Dual-Gene Reporter-Amplifier Architecture for Enhancing the Sensitivity of Molecular MRI by Water Exchange.
- Author
-
Huang, Yimeng, Chen, Xinyue, Zhu, Ziyue, and Mukherjee, Arnab
- Subjects
MRI ,Oatp1 ,aquaporins ,diffusion ,reporter genes ,Water ,Humans ,Magnetic Resonance Imaging ,Aquaporin 1 ,Genes ,Reporter ,Solute Carrier Organic Anion Transporter Family Member 1B3 ,Gadolinium ,Contrast Media ,HEK293 Cells ,Animals - Abstract
The development of genetic reporters for magnetic resonance imaging (MRI) is essential for investigating biological functions in vivo. However, current MRI reporters have low sensitivity, making it challenging to create significant contrast against the tissue background, especially when only a small fraction of cells express the reporter. To overcome this limitation, we developed an approach for amplifying the sensitivity of molecular MRI by combining a chemogenetic contrast mechanism with a biophysical approach to increase water diffusion through the co-expression of a dual-gene construct comprising an organic anion transporting polypeptide, Oatp1b3, and a water channel, Aqp1. We first show that the expression of Aqp1 amplifies MRI contrast in cultured cells engineered to express Oatp1b3. We demonstrate that the contrast amplification is caused by Aqp1-driven increase in water exchange, which provides the gadolinium ions internalized by Oatp1b3-expressing cells with access to a larger water pool compared with exchange-limited conditions. We further show that our methodology allows cells to be detected using approximately 10-fold lower concentrations of gadolinium than that in the Aqp1-free scenario. Finally, we show that our approach enables the imaging of mixed-cell cultures containing a low fraction of Oatp1b3-labeled cells that are undetectable on the basis of Oatp1b3 expression alone.
- Published
- 2024
48. An Inner Mitochondrial Membrane Microprotein from the SLC35A4 Upstream ORF Regulates Cellular Metabolism.
- Author
-
Rocha, Andréa, Pai, Victor, Perkins, Guy, Chang, Tina, Ma, Jiao, De Souza, Eduardo, Chu, Qian, Vaughan, Joan, Diedrich, Jolene, Ellisman, Mark, and Saghatelian, Alan
- Subjects
cellular metabolism ,inner mitochondrial membrane ,microprotein ,mitochondria ,upstream open reading frame (uORF) ,Humans ,5 Untranslated Regions ,Amino Acid Sequence ,Mitochondria ,Mitochondrial Membranes ,Mitochondrial Proteins ,Open Reading Frames ,Protein Biosynthesis ,RNA ,Messenger ,Nucleotide Transport Proteins ,HEK293 Cells - Abstract
Upstream open reading frames (uORFs) are cis-acting elements that can dynamically regulate the translation of downstream ORFs by suppressing downstream translation under basal conditions and, in some cases, increasing downstream translation under stress conditions. Computational and empirical methods have identified uORFs in the 5-UTRs of approximately half of all mouse and human transcripts, making uORFs one of the largest regulatory elements known. Because the prevailing dogma was that eukaryotic mRNAs produce a single functional protein, the peptides and small proteins, or microproteins, encoded by uORFs were rarely studied. We hypothesized that a uORF in the SLC35A4 mRNA is producing a functional microprotein (SLC35A4-MP) because of its conserved amino acid sequence. Through a series of biochemical and cellular experiments, we find that the 103-amino acid SLC35A4-MP is a single-pass transmembrane inner mitochondrial membrane (IMM) microprotein. The IMM contains the protein machinery crucial for cellular respiration and ATP generation, and loss of function studies with SLC35A4-MP significantly diminish maximal cellular respiration, indicating a vital role for this microprotein in cellular metabolism. The findings add SLC35A4-MP to the growing list of functional microproteins and, more generally, indicate that uORFs that encode conserved microproteins are an untapped reservoir of functional microproteins.
- Published
- 2024
49. Analysis of Tumor-Associated AXIN1 Missense Mutations Identifies Variants That Activate β-Catenin Signaling.
- Author
-
Peppelenbosch, Maikel, Lebbink, Joyce, Smits, Ron, Zhang, Ruyi, Li, Shanshan, Schippers, Kelly, Li, Yunlong, Eimers, Boaz, Lavrijsen, Marla, Wang, Ling, Cui, Guofei, and Chen, Xin
- Subjects
Axin Protein ,Humans ,beta Catenin ,Mutation ,Missense ,Signal Transduction ,Glycogen Synthase Kinase 3 beta ,Liver Neoplasms ,Neoplasms ,HEK293 Cells ,Cell Line ,Tumor ,Protein Binding - Abstract
UNLABELLED: AXIN1 is a major component of the β-catenin destruction complex and is frequently mutated in various cancer types, particularly liver cancers. Truncating AXIN1 mutations are recognized to encode a defective protein that leads to β-catenin stabilization, but the functional consequences of missense mutations are not well characterized. Here, we first identified the GSK3β, β-catenin, and RGS/APC interaction domains of AXIN1 that are the most critical for proper β-catenin regulation. Analysis of 80 tumor-associated variants in these domains identified 18 that significantly affected β-catenin signaling. Coimmunoprecipitation experiments revealed that most of them lost binding to the binding partner corresponding to the mutated domain. A comprehensive protein structure analysis predicted the consequences of these mutations, which largely overlapped with the observed effects on β-catenin signaling in functional experiments. The structure analysis also predicted that loss-of-function mutations within the RGS/APC interaction domain either directly affected the interface for APC binding or were located within the hydrophobic core and destabilized the entire structure. In addition, truncated AXIN1 length inversely correlated with the β-catenin regulatory function, with longer proteins retaining more functionality. These analyses suggest that all AXIN1-truncating mutations at least partially affect β-catenin regulation, whereas this is only the case for a subset of missense mutations. Consistently, most colorectal and liver cancers carrying missense variants acquire mutations in other β-catenin regulatory genes such as APC and CTNNB1. These results will aid the functional annotation of AXIN1 mutations identified in large-scale sequencing efforts or in individual patients. SIGNIFICANCE: Characterization of 80 tumor-associated missense variants of AXIN1 reveals a subset of 18 mutations that disrupt its β-catenin regulatory function, whereas the majority are passenger mutations.
- Published
- 2024
50. Selective targeting of mu opioid receptors to primary cilia
- Author
-
Fagan, Rita R, Lee, David F, Geron, Matan, Scherrer, Grégory, von Zastrow, Mark, and Ehrlich, Aliza T
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Neurosciences ,Substance Misuse ,Drug Abuse (NIDA only) ,Opioids ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Generic health relevance ,Receptors ,Opioid ,mu ,Cilia ,Animals ,Mice ,Humans ,HEK293 Cells ,Protein Transport ,CP: Cell biology ,G protein-coupled receptor ,MOR ,TULP3 ,brain ,mouse ,neuronal ,primary cilium ,recycling ,GPCR ,NEURONAL ,Medical Physiology ,Biological sciences - Abstract
Opioid receptors are therapeutically important G protein-coupled receptors (GPCRs) with diverse neuromodulatory effects. The functional consequences of opioid receptor activation are known to depend on receptor location in the plasma membrane, but mechanisms mediating selective localization of receptors to any particular membrane domain remain elusive. Here, we demonstrate the targeting of the mu opioid receptor (MOR) to the primary cilium, a discrete microdomain of the somatic plasma membrane, both in vivo and in cultured cells. We further show that ciliary targeting is specific to MORs, requires a 17-residue sequence unique to the MOR cytoplasmic tail, and additionally requires the Tubby-like protein 3 (TULP3) ciliary adaptor protein. Our results reveal the potential for opioid receptors to undergo selective localization to the primary cilium. We propose that ciliary targeting is mediated through an elaboration of the recycling pathway, directed by a specific C-terminal recycling sequence in cis and requiring TULP3 in trans.
- Published
- 2024
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.