34 results on '"Butty, Vincent L."'
Search Results
2. Single-cell transcriptomic profiling of the aging mouse brain
- Author
-
Ximerakis, Methodios, Lipnick, Scott L., Innes, Brendan T., Simmons, Sean K., Adiconis, Xian, Dionne, Danielle, Mayweather, Brittany A., Nguyen, Lan, Niziolek, Zachary, Ozek, Ceren, Butty, Vincent L., Isserlin, Ruth, Buchanan, Sean M., Levine, Stuart S., Regev, Aviv, Bader, Gary D., Levin, Joshua Z., and Rubin, Lee L.
- Published
- 2019
- Full Text
- View/download PDF
3. Digital Genotyping and Haplotyping with Polymerase Colonies
- Author
-
Mitra, Robi D., Butty, Vincent L., Shendure, Jay, Williams, Benjamin R., Housman, David E., and Church, George M.
- Published
- 2003
4. Cross-site comparison of ribosomal depletion kits for Illumina RNAseq library construction
- Author
-
Herbert, Zachary T., Kershner, Jamie P., Butty, Vincent L., Thimmapuram, Jyothi, Choudhari, Sulbha, Alekseyev, Yuriy O., Fan, Jun, Podnar, Jessica W., Wilcox, Edward, Gipson, Jenny, Gillaspy, Allison, Jepsen, Kristen, BonDurant, Sandra Splinter, Morris, Krystalynne, Berkeley, Maura, LeClerc, Ashley, Simpson, Stephen D., Sommerville, Gary, Grimmett, Leslie, Adams, Marie, and Levine, Stuart S.
- Published
- 2018
- Full Text
- View/download PDF
5. The endoplasmic reticulum proteostasis network profoundly shapes the protein sequence space accessible to HIV envelope.
- Author
-
Yoon, Jimin, Nekongo, Emmanuel E., Patrick, Jessica E., Hui, Tiffani, Phillips, Angela M., Ponomarenko, Anna I., Hendel, Samuel J., Sebastian, Rebecca M., Zhang, Yu Meng, Butty, Vincent L., Ogbunugafor, C. Brandon, Lin, Yu-Shan, and Shoulders, Matthew D.
- Subjects
SEQUENCE spaces ,AMINO acid sequence ,ENDOPLASMIC reticulum ,UNFOLDED protein response ,IMMUNOGLOBULINS ,HIV - Abstract
The sequence space accessible to evolving proteins can be enhanced by cellular chaperones that assist biophysically defective clients in navigating complex folding landscapes. It is also possible, at least in theory, for proteostasis mechanisms that promote strict quality control to greatly constrain accessible protein sequence space. Unfortunately, most efforts to understand how proteostasis mechanisms influence evolution rely on artificial inhibition or genetic knockdown of specific chaperones. The few experiments that perturb quality control pathways also generally modulate the levels of only individual quality control factors. Here, we use chemical genetic strategies to tune proteostasis networks via natural stress response pathways that regulate the levels of entire suites of chaperones and quality control mechanisms. Specifically, we upregulate the unfolded protein response (UPR) to test the hypothesis that the host endoplasmic reticulum (ER) proteostasis network shapes the sequence space accessible to human immunodeficiency virus-1 (HIV-1) envelope (Env) protein. Elucidating factors that enhance or constrain Env sequence space is critical because Env evolves extremely rapidly, yielding HIV strains with antibody- and drug-escape mutations. We find that UPR-mediated upregulation of ER proteostasis factors, particularly those controlled by the IRE1-XBP1s UPR arm, globally reduces Env mutational tolerance. Conserved, functionally important Env regions exhibit the largest decreases in mutational tolerance upon XBP1s induction. Our data indicate that this phenomenon likely reflects strict quality control endowed by XBP1s-mediated remodeling of the ER proteostasis environment. Intriguingly, and in contrast, specific regions of Env, including regions targeted by broadly neutralizing antibodies, display enhanced mutational tolerance when XBP1s is induced, hinting at a role for host proteostasis network hijacking in potentiating antibody escape. These observations reveal a key function for proteostasis networks in decreasing instead of expanding the sequence space accessible to client proteins, while also demonstrating that the host ER proteostasis network profoundly shapes the mutational tolerance of Env in ways that could have important consequences for HIV adaptation. The host cell's endoplasmic reticulum proteostasis network has a profound, constraining impact on the protein sequence space accessible to HIV's envelope protein, which is a major target of the host's adaptive immune system; in particular, upregulation of stringent quality control pathways appears to restrict the viability of destabilizing envelope variants. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
6. High-throughput Minitaturized RNA-Seq Library Preparation.
- Author
-
Mildrum, Samuel, Hendricks, Austin, Stortchevoi, Alexei, Kamelamela, Noelani, Butty, Vincent L., and Levine, Stuart S.
- Published
- 2020
- Full Text
- View/download PDF
7. HSF1 Activation Can Restrict HIV Replication.
- Author
-
Nekongo, Emmanuel E., Ponomarenko, Anna I., Dewal, Mahender B., Butty, Vincent L., Browne, Edward P., and Shoulders, Matthew D.
- Published
- 2020
- Full Text
- View/download PDF
8. H3K27me3-mediated silencing of structural genes is required for zebrafish heart regeneration.
- Author
-
Ben-Yair, Raz, Butty, Vincent L., Busby, Michele, Yutong Qiu, Levine, Stuart S., Goren, Alon, Boyer, Laurie A., Burns, C. Geoffrey, and Burns, Caroline E.
- Subjects
- *
CARDIAC regeneration , *GENE silencing , *GENETIC regulation , *CELL cycle , *GENE expression , *CHROMATIN , *BONE regeneration - Abstract
Deciphering the genetic and epigenetic regulation of cardiomyocyte proliferation in organisms, such as zebrafish, capable of robust cardiac renewal represents an attractive inroad towards regenerating the human heart. Using integrated highthroughput transcriptional and chromatin analyses, we identified a strong association between H3K27me3 deposition and reduced sarcomere and cytoskeletal gene expression in proliferative cardiomyocytes following injury. To move beyond an association, we generated an inducible transgenic strain expressing a mutant version of histone 3, H3.3K27M that inhibits H3K27me3 catalysis in cardiomyocytes during the regenerative window. Hearts comprised of H3.3K27M-expressing cardiomyocytes fail to regenerate with wound edge cells showing heightened expression of structural genes and prominent sarcomere structures. Although cell cycle re-entry was unperturbed, cytokinesis and wound invasion were significantly compromised. Collectively, our study identifies H3K27me3-mediated silencing of structural genes as requisite for zebrafish heart regeneration and suggests that repression of similar structural components in the border zone of infarcted human hearts might improve its regenerative capacity. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
9. Global transcriptional regulation of innate immunity by ATF-7 in C. elegans.
- Author
-
Fletcher, Marissa, Tillman, Erik J., Butty, Vincent L., Levine, Stuart S., and Kim, Dennis H.
- Subjects
CAENORHABDITIS elegans ,NATURAL immunity ,PSEUDOMONAS aeruginosa ,IMMUNOPRECIPITATION ,RNA sequencing ,TRANSCRIPTION factors ,CAENORHABDITIS - Abstract
The nematode Caenorhabditis elegans has emerged as a genetically tractable animal host in which to study evolutionarily conserved mechanisms of innate immune signaling. We previously showed that the PMK-1 p38 mitogen-activated protein kinase (MAPK) pathway regulates innate immunity of C. elegans through phosphorylation of the CREB/ATF bZIP transcription factor, ATF-7. Here, we have undertaken a genomic analysis of the transcriptional response of C. elegans to infection by Pseudomonas aeruginosa, combining genome-wide expression analysis by RNA-seq with ATF-7 chromatin immunoprecipitation followed by sequencing (ChIP-Seq). We observe that PMK-1-ATF-7 activity regulates a majority of all genes induced by pathogen infection, and observe ATF-7 occupancy in regulatory regions of pathogen-induced genes in a PMK-1-dependent manner. Moreover, functional analysis of a subset of these ATF-7-regulated pathogen-induced target genes supports a direct role for this transcriptional response in host defense. The genome-wide regulation through PMK-1– ATF-7 signaling reveals a striking level of control over the innate immune response to infection through a single transcriptional regulator. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
10. An Adaptable Platform for Directed Evolution in Human Cells.
- Author
-
Berman, Chet M., Papa III, Louis J., Hendel, Samuel J., Moore, Christopher L., Suen, Patreece H., Weickhardt, Alexander F., Doan, Ngoc-Duc, Kumar, Caiden M., Uil, Taco G., Butty, Vincent L., Hoeben, Robert C., and Shoulders, Matthew D.
- Published
- 2018
- Full Text
- View/download PDF
11. Destabilized adaptive influenza variants critical for innate immune system escape are potentiated by host chaperones.
- Author
-
Phillips, Angela M., Ponomarenko, Anna I., Chen, Kenny, Ashenberg, Orr, Miao, Jiayuan, McHugh, Sean M., Butty, Vincent L., Whittaker, Charles A., Moore, Christopher L., Bloom, Jesse D., Lin, Yu-Shan, and Shoulders, Matthew D.
- Subjects
INFLUENZA ,IMMUNE system ,MOLECULAR chaperones ,VIRUSES ,NUCLEOPROTEIN genetics - Abstract
The threat of viral pandemics demands a comprehensive understanding of evolution at the host–pathogen interface. Here, we show that the accessibility of adaptive mutations in influenza nucleoprotein at fever-like temperatures is mediated by host chaperones. Particularly noteworthy, we observe that the Pro283 nucleoprotein variant, which (1) is conserved across human influenza strains, (2) confers resistance to the Myxovirus resistance protein A (MxA) restriction factor, and (3) critically contributed to adaptation to humans in the 1918 pandemic influenza strain, is rendered unfit by heat shock factor 1 inhibition–mediated host chaperone depletion at febrile temperatures. This fitness loss is due to biophysical defects that chaperones are unavailable to address when heat shock factor 1 is inhibited. Thus, influenza subverts host chaperones to uncouple the biophysically deleterious consequences of viral protein variants from the benefits of immune escape. In summary, host proteostasis plays a central role in shaping influenza adaptation, with implications for the evolution of other viruses, for viral host switching, and for antiviral drug development. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
12. A Method for Selective Depletion of Zn(II) Ions from Complex Biological Media and Evaluation of Cellular Consequences of Zn(II) Deficiency.
- Author
-
Richardson, Christopher E. R., Cunden, Lisa S., Nolan, Elizabeth M., Lippard, Stephen J., Shoulders, Matthew D., and Butty, Vincent L.
- Published
- 2018
- Full Text
- View/download PDF
13. Alternative RNA splicing in the endothelium mediated in part by Rbfox2 regulates the arterial response to low flow.
- Author
-
Murphy, Patrick A., Butty, Vincent L., Boutz, Paul L., Begum, Shahinoor, Kimble, Amy L., Sharp, Phillip A., Burge, Christopher B., and Hynes, Richard O.
- Subjects
- *
ALTERNATIVE RNA splicing , *ENDOTHELIUM , *RNA-binding proteins , *BLOOD flow , *ATHEROSCLEROSIS - Abstract
Low and disturbed blood flow drives the progression of arterial diseases including atherosclerosis and aneurysms. The endothelial response to flow and its interactions with recruited platelets and leukocytes determine disease progression. Here, we report widespread changes in alternative splicing of pre-mRNA in the flow-activated murine arterial endothelium in vivo. Alternative splicing was suppressed by depletion of platelets and macrophages recruited to the arterial endothelium under low and disturbed flow. Binding motifs for the Rbfox-family are enriched adjacent to many of the regulated exons. Endothelial deletion of Rbfox2, the only family member expressed in arterial endothelium, suppresses a subset of the changes in transcription and RNA splicing induced by low flow. Our data reveal an alternative splicing program activated by Rbfox2 in the endothelium on recruitment of platelets and macrophages and demonstrate its relevance in transcriptional responses during flow-driven vascular inflammation. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
14. Host proteostasis modulates influenza evolution.
- Author
-
Phillips, Angela M., Gonzalez, Luna O., Nekongo, Emmanuel E., Ponomarenko, Anna I., McHugh, Sean M., Butty, Vincent L., Levine, Stuart S., Yu-Shan Lin, Mirny, Leonid A., and Shoulders, Matthew D.
- Published
- 2017
- Full Text
- View/download PDF
15. Expanded MutaT7 toolkit efficiently and simultaneously accesses all possible transition mutations in bacteria.
- Author
-
Mengiste, Amanuella A, Wilson, Robert H, Weissman, Rachel F, Papa III, Louis J, Hendel, Samuel J, Moore, Christopher L, Butty, Vincent L, and Shoulders, Matthew D
- Published
- 2023
- Full Text
- View/download PDF
16. Transcriptional Reversion of Cardiac Myocyte Fate During Mammalian Cardiac Regeneration.
- Author
-
O'Meara, Caitlin C., Wamstad, Joseph A., Gladstone, Rachel A., Fomovsky, Gregory M., Butty, Vincent L., Shrikumar, Avanti, Gannon, Joseph B., Boyer, Laurie A., and Lee, Richard T.
- Published
- 2015
- Full Text
- View/download PDF
17. A single-cell liver atlas of Plasmodium vivax infection.
- Author
-
Mancio-Silva, Liliana, Gural, Nil, Real, Eliana, Wadsworth II, Marc H., Butty, Vincent L., March, Sandra, Nerurkar, Niketa, Hughes, Travis K., Roobsoong, Wanlapa, Fleming, Heather E., Whittaker, Charlie A., Levine, Stuart S., Sattabongkot, Jetsumon, Shalek, Alex K., and Bhatia, Sangeeta N.
- Abstract
Malaria-causing Plasmodium vivax parasites can linger in the human liver for weeks to years and reactivate to cause recurrent blood-stage infection. Although they are an important target for malaria eradication, little is known about the molecular features of replicative and non-replicative intracellular liver-stage parasites and their host cell dependence. Here, we leverage a bioengineered human microliver platform to culture patient-derived P. vivax parasites for transcriptional profiling. Coupling enrichment strategies with bulk and single-cell analyses, we capture both parasite and host transcripts in individual hepatocytes throughout the course of infection. We define host- and state-dependent transcriptional signatures and identify unappreciated populations of replicative and non-replicative parasites that share features with sexual transmissive forms. We find that infection suppresses the transcription of key hepatocyte function genes and elicits an anti-parasite innate immune response. Our work provides a foundation for understanding host-parasite interactions and reveals insights into the biology of P. vivax dormancy and transmission. [Display omitted] • MPCC and Seq-Well platforms enable single-cell survey of P. vivax liver infection • Dual transcriptome analysis links dormancy and sexual conversion to host state • Single-cell profiling reveals dynamic host responses in infected and bystander cells • Dataset is validated via in situ transcript detection and functional assays Plasmodium vivax parasites cause relapsing human malaria, which is considered a barrier to its eradication. Mancio-Silva et al. perform time-resolved, dual single-cell transcriptional profiling of this elusive organism to garner insights into hypnozoite dormancy and activation, hepatic sexual commitment, and host immune response to infection. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
18. Polycomb Repressive Complex 2 Regulates Lineage Fidelity during Embryonic Stem Cell Differentiation.
- Author
-
Thornton, Seraphim R., Butty, Vincent L., Levine, Stuart S., and Boyer, Laurie A.
- Subjects
- *
HISTONES , *CELL differentiation , *EMBRYONIC stem cells , *POLYCOMB group proteins , *DNA methylation - Abstract
Polycomb Repressive Complex 2 (PRC2) catalyzes histone H3 lysine 27 tri-methylation (H3K27me3), an epigenetic modification associated with gene repression. H3K27me3 is enriched at the promoters of a large cohort of developmental genes in embryonic stem cells (ESCs). Loss of H3K27me3 leads to a failure of ESCs to properly differentiate, making it difficult to determine the precise roles of PRC2 during lineage commitment. Moreover, while studies suggest that PRC2 prevents DNA methylation, how these two epigenetic regulators coordinate to regulate lineage programs is poorly understood. Using several PRC2 mutant ESC lines that maintain varying levels of H3K27me3, we found that partial maintenance of H3K27me3 allowed for proper temporal activation of lineage genes during directed differentiation of ESCs to spinal motor neurons (SMNs). In contrast, genes that function to specify other lineages failed to be repressed in these cells, suggesting that PRC2 is also necessary for lineage fidelity. We also found that loss of H3K27me3 leads to a modest gain in DNA methylation at PRC2 target regions in both ESCs and in SMNs. Our study demonstrates a critical role for PRC2 in safeguarding lineage decisions and in protecting genes against inappropriate DNA methylation. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
19. Multiplexed DNA repair assays for multiple lesions and multiple doses via transcription inhibition and transcriptional mutagenesis.
- Author
-
Nagel, Zachary D., Margulies, Carrie M., Chaim, Isaac A., McRee, Siobhan K., Mazzucato, Patrizia, Ahmad, Anwaar, Abo, Ryan P., Butty, Vincent L., Forget, Anthony L., and Samson, Leona D.
- Subjects
DNA repair ,DNA damage ,FLUORESCENCE ,RNA polymerases ,DISEASE susceptibility - Abstract
The capacity to repair different types of DNA damage varies among individuals, making them more or less susceptible to the detrimental health consequences of damage exposures. Current methods for measuring DNA repair capacity (DRC) are relatively labor intensive, often indirect, and usually limited to a single repair pathway. Here, we describe a fluorescence-based multiplex flow-cytometric host cell reactivation assay (FM-HCR) that measures the ability of human cells to repair plasmid reporters, each bearing a different type of DNA damage or different doses of the same type of DNA damage. FM-HCR simultaneously measures repair capacity in any four of the following pathways: nucleotide excision repair, mismatch repair, base excision repair, nonhomologous end joining, homologous recombination, and methylguanine methyltransferase. We show that FM-HCR can measure interindividual DRC differences in a panel of 24 cell lines derived from genetically diverse, apparently healthy individuals, and we show that FM-HCR may be used to identify inhibitors or enhancers of DRC. We further develop a next-generation sequencing-based HCR assay (HCR-Seq) that detects rare transcriptional mutagenesis events due to lesion bypass by RNA polymerase, providing an added dimension to DRC measurements. FM-HCR and HCR-Seq provide powerful tools for exploring relationships among global DRC, disease susceptibility, and optimal treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
20. H2A.Z Acidic Patch Couples Chromatin Dynamics to Regulation of Gene Expression Programs during ESC Differentiation.
- Author
-
Subramanian, Vidya, Mazumder, Aprotim, Surface, Lauren E., Butty, Vincent L., Fields, Paul A., Alwan, Allison, Torrey, Lillian, Thai, Kevin K., Levine, Stuart S., Bathe, Mark, and Boyer, Laurie A.
- Subjects
HISTONES ,EMBRYOLOGY ,EMBRYONIC stem cell research ,GENE expression ,CHROMATIN - Abstract
The histone H2A variant H2A.Z is essential for embryonic development and for proper control of developmental gene expression programs in embryonic stem cells (ESCs). Divergent regions of amino acid sequence of H2A.Z likely determine its functional specialization compared to core histone H2A. For example, H2A.Z contains three divergent residues in the essential C-terminal acidic patch that reside on the surface of the histone octamer as an uninterrupted acidic patch domain; however, we know little about how these residues contribute to chromatin structure and function. Here, we show that the divergent amino acids Gly92, Asp97, and Ser98 in the H2A.Z C-terminal acidic patch (H2A.Z
AP3 ) are critical for lineage commitment during ESC differentiation. H2A.Z is enriched at most H3K4me3 promoters in ESCs including poised, bivalent promoters that harbor both activating and repressive marks, H3K4me3 and H3K27me3 respectively. We found that while H2A.ZAP3 interacted with its deposition complex and displayed a highly similar distribution pattern compared to wild-type H2A.Z, its enrichment levels were reduced at target promoters. Further analysis revealed that H2A.ZAP3 was less tightly associated with chromatin, suggesting that the mutant is more dynamic. Notably, bivalent genes in H2A.ZAP3 ESCs displayed significant changes in expression compared to active genes. Moreover, bivalent genes in H2A.ZAP3 ESCs gained H3.3, a variant associated with higher nucleosome turnover, compared to wild-type H2A.Z. We next performed single cell imaging to measure H2A.Z dynamics. We found that H2A.ZAP3 displayed higher mobility in chromatin compared to wild-type H2A.Z by fluorescent recovery after photobleaching (FRAP). Moreover, ESCs treated with the transcriptional inhibitor flavopiridol resulted in a decrease in the H2A.ZAP3 mobile fraction and an increase in its occupancy at target genes indicating that the mutant can be properly incorporated into chromatin. Collectively, our work suggests that the divergent residues in the H2A.Z acidic patch comprise a unique domain that couples control of chromatin dynamics to the regulation of developmental gene expression patterns during lineage commitment. [ABSTRACT FROM AUTHOR]- Published
- 2013
- Full Text
- View/download PDF
21. Braveheart, a Long Noncoding RNA Required for Cardiovascular Lineage Commitment
- Author
-
Klattenhoff, Carla A., Scheuermann, Johanna C., Surface, Lauren E., Bradley, Robert K., Fields, Paul A., Steinhauser, Matthew L., Ding, Huiming, Butty, Vincent L., Torrey, Lillian, Haas, Simon, Abo, Ryan, Tabebordbar, Mohammadsharif, Lee, Richard Theodore, Burge, Christopher B., and Boyer, Laurie A.
- Abstract
Long noncoding RNAs (lncRNAs) are often expressed in a development-specific manner, yet little is known about their roles in lineage commitment. Here, we identified Braveheart (Bvht), a heart-associated lncRNA in mouse. Using multiple embryonic stem cell (ESC) differentiation strategies, we show that Bvht is required for progression of nascent mesoderm toward a cardiac fate. We find that Bvht is necessary for activation of a core cardiovascular gene network and functions upstream of mesoderm posterior 1 (MesP1), a master regulator of a common multipotent cardiovascular progenitor. We also show that Bvht interacts with SUZ12, a component of polycomb-repressive complex 2 (PRC2), during cardiomyocyte differentiation, suggesting that Bvht mediates epigenetic regulation of cardiac commitment. Finally, we demonstrate a role for Bvht in maintaining cardiac fate in neonatal cardiomyocytes. Together, our work provides evidence for a long noncoding RNA with critical roles in the establishment of the cardiovascular lineage during mammalian development., Stem Cell and Regenerative Biology
- Published
- 2013
- Full Text
- View/download PDF
22. Failed Progenitor Specification Underlies the Cardiopharyngeal Phenotypes in a Zebrafish Model of 22q11.2 Deletion Syndrome.
- Author
-
Guner-Ataman, Burcu, González-Rosa, Juan Manuel, Shah, Harsh N., Butty, Vincent L., Jeffrey, Spencer, Abrial, Maryline, Boyer, Laurie A., Burns, C. Geoffrey, and Burns, Caroline E.
- Abstract
Summary Microdeletions involving TBX1 result in variable congenital malformations known collectively as 22q11.2 deletion syndrome (22q11.2DS). Tbx1-deficient mice and zebrafish recapitulate several disease phenotypes, including pharyngeal arch artery (PAA), head muscle (HM), and cardiac outflow tract (OFT) deficiencies. In zebrafish, these structures arise from nkx2.5 + progenitors in pharyngeal arches 2 – 6. Because pharyngeal arch morphogenesis is compromised in Tbx1-deficient animals, the malformations were considered secondary. Here, we report that the PAA, HM, and OFT phenotypes in tbx1 mutant zebrafish are primary and arise prior to pharyngeal arch morphogenesis from failed specification of the nkx2.5 + pharyngeal lineage. Through in situ analysis and lineage tracing, we reveal that nkx2.5 and tbx1 are co-expressed in this progenitor population. Furthermore, we present evidence suggesting that gdf3 -ALK4 signaling is a downstream mediator of nkx2.5 + pharyngeal lineage specification. Collectively, these studies support a cellular mechanism potentially underlying the cardiovascular and craniofacial defects observed in the 22q11.2DS population. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
23. In Vitro Culture, Drug Sensitivity, and Transcriptome of Plasmodium Vivax Hypnozoites.
- Author
-
Gural, Nil, Mancio-Silva, Liliana, Miller, Alex B., Galstian, Ani, Butty, Vincent L., Levine, Stuart S., Patrapuvich, Rapatbhorn, Desai, Salil P., Mikolajczak, Sebastian A., Kappe, Stefan H.I., Fleming, Heather E., March, Sandra, Sattabongkot, Jetsumon, and Bhatia, Sangeeta N.
- Abstract
Summary The unique relapsing nature of Plasmodium vivax infection is a major barrier to malaria eradication. Upon infection, dormant liver-stage forms, hypnozoites, linger for weeks to months and then relapse to cause recurrent blood-stage infection. Very little is known about hypnozoite biology; definitive biomarkers are lacking and in vitro platforms that support phenotypic studies are needed. Here, we recapitulate the entire liver stage of P. vivax in vitro , using a multiwell format that incorporates micropatterned primary human hepatocyte co-cultures (MPCCs). MPCCs feature key aspects of P. vivax biology, including establishment of persistent small forms and growing schizonts, merosome release, and subsequent infection of reticulocytes. We find that the small forms exhibit previously described hallmarks of hypnozoites, and we pilot MPCCs as a tool for testing candidate anti-hypnozoite drugs. Finally, we employ a hybrid capture strategy and RNA sequencing to describe the hypnozoite transcriptome and gain insight into its biology. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
24. Geometry-dependent functional changes in iPSC-derived cardiomyocytes probed by functional imaging and RNA sequencing
- Author
-
Aviv Regev, Vincent L. Butty, B. Alexander Yi, Miao-Ping Chien, Karthik Shekhar, Christopher A. Werley, Laurie A. Boyer, Jellert Gaublomme, Joel M. Kralj, William Bloxham, Adam E. Cohen, Massachusetts Institute of Technology. Department of Biological Engineering, Massachusetts Institute of Technology. Department of Biology, Koch Institute for Integrative Cancer Research at MIT, Butty, Vincent L G, Boyer, Laurie Ann, Regev, Aviv, and Pesce, Maurizio
- Subjects
0301 basic medicine ,Transcription, Genetic ,Physiology ,Molecular biology ,Cellular differentiation ,lcsh:Medicine ,Gene Expression ,Action Potentials ,Geometry ,Cardiovascular ,Sequencing techniques ,Gene expression ,Medicine and Health Sciences ,Myocyte ,Myocytes, Cardiac ,Induced pluripotent stem cell ,lcsh:Science ,Cells, Cultured ,Multidisciplinary ,Cultured ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Cardiac electrophysiology ,RNA sequencing ,Cell Differentiation ,Calcium Imaging ,3. Good health ,Cell biology ,Electrophysiology ,Heart Disease ,Bioassays and Physiological Analysis ,Biological Cultures ,Cardiac ,Sequence Analysis ,Transcription ,Research Article ,General Science & Technology ,Imaging Techniques ,Cells ,1.1 Normal biological development and functioning ,Induced Pluripotent Stem Cells ,Neurophysiology ,Neuroimaging ,Biology ,Research and Analysis Methods ,Membrane Potential ,03 medical and health sciences ,Calcium imaging ,Genetic ,Underpinning research ,Fluorescence Imaging ,Genetics ,Humans ,Stem Cell Research - Embryonic - Human ,Myocytes ,Stem Cell Research - Induced Pluripotent Stem Cell ,Sequence Analysis, RNA ,lcsh:R ,Electrophysiological Techniques ,Biology and Life Sciences ,Cell Cultures ,Stem Cell Research ,Embryonic stem cell ,Electrophysiological Phenomena ,030104 developmental biology ,Molecular biology techniques ,Biophysics ,RNA ,lcsh:Q ,Calcium ,Cardiac Electrophysiology ,Neuroscience - Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) are a promising platform for cardiac studies in vitro, and possibly for tissue repair in humans. However, hiPSC-CM cells tend to retain morphology, metabolism, patterns of gene expression, and electrophysiology similar to that of embryonic cardiomyocytes. We grew hiPSC-CM in patterned islands of different sizes and shapes, and measured the effect of island geometry on action potential waveform and calcium dynamics using optical recordings of voltage and calcium from 970 islands of different sizes. hiPSC-CM in larger islands showed electrical and calcium dynamics indicative of greater functional maturity. We then compared transcriptional signatures of the small and large islands against a developmental time course of cardiac differentiation. Although island size had little effect on expression of most genes whose levels differed between hiPSC-CM and adult primary CM, we identified a subset of genes for which island size drove the majority (58%) of the changes associated with functional maturation. Finally, we patterned hiPSC-CM on islands with a variety of shapes to probe the relative contributions of soluble factors, electrical coupling, and direct cell-cell contacts to the functional maturation. Collectively, our data show that optical electrophysiology is a powerful tool for assaying hiPSC-CM maturation, and that island size powerfully drives activation of a subset of genes involved in cardiac maturation, National Science Foundation (U.S.). Center on Emergent Behaviors of Integrated Cellular Systems (CBET-0939511), National Cancer Institute (U.S.) (BioMicro Center. Award P30-CA14051), Howard Hughes Medical Institute
- Published
- 2016
25. Failed Progenitor Specification Underlies the Cardiopharyngeal Phenotypesin a Zebrafish Model of 22q11.2 Deletion Syndrome
- Author
-
Laurie A. Boyer, Caroline E. Burns, Burcu Guner-Ataman, Harsh N. Shah, C. Geoffrey Burns, Vincent L. Butty, Spencer Jeffrey, Juan Manuel González-Rosa, Maryline Abrial, Massachusetts Institute of Technology. Department of Biology, Butty, Vincent L G, and Boyer, Laurie Ann
- Subjects
0301 basic medicine ,TBX1 ,Lineage (genetic) ,22q11 Deletion Syndrome ,Population ,Morphogenesis ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,stomatognathic system ,medicine ,Animals ,Cell Lineage ,Progenitor cell ,education ,Zebrafish ,Embryonic Stem Cells ,Progenitor ,education.field_of_study ,Cell Differentiation ,Zebrafish Proteins ,biology.organism_classification ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Phenotype ,embryonic structures ,Homeobox Protein Nkx-2.5 ,Pharynx ,T-Box Domain Proteins ,Pharyngeal arch - Abstract
SUMMARY Microdeletions involving TBX1 result in variable congenital malformations known collectively as 22q11.2 deletion syndrome (22q11.2DS). Tbx1-deficient mice and zebrafish recapitulate several disease phenotypes, including pharyngeal arch artery (PAA), head muscle (HM), and cardiac outflow tract (OFT) deficiencies. In zebrafish, these structures arise from nkx2.5+ progenitors in pharyngeal arches 2–6. Because pharyngeal arch morphogenesis is compromised in Tbx1-deficient animals, the malformations were considered secondary. Here, we report that the PAA, HM, and OFT phenotypes in tbx1 mutant zebrafish are primary and arise prior to pharyngeal arch morphogenesis from failed specification of the nkx2.5+ pharyngeal lineage. Through in situ analysis and lineage tracing, we reveal that nkx2.5 and tbx1 are co-expressed in this progenitor population. Furthermore, we present evidence suggesting that gdf3-ALK4 signaling is a downstream mediator of nkx2.5+ pharyngeal lineage specification. Collectively, these studies support a cellular mechanism potentially underlying the cardiovascular and cranio-facial defects observed in the 22q11.2DS population., In Brief Microdeletions encompassing the TBX1 locus cause 22q11.2 deletion syndrome (DS), which is characterized by congenital heart, aorta, and craniofacial malformations. Using a zebrafish model of 22q11.2DS, Guner-Ataman et al. demonstrate that tbx1-mutant animals fail to specify the nkx2.5+ progenitor population that gives rise to the affected structures., Graphical Abstract
- Published
- 2018
26. Human cartilage model of the precocious osteoarthritis-inducing COL2A1 p.Arg719Cys reveals pathology-driving matrix defects and a failure of the ER proteostasis network to recognize the defective procollagen-II.
- Author
-
Yammine KM, Abularach SM, Xiong M, Kim SY, Bikovtseva AA, Butty VL, Schiavoni RP, Bateman JF, Lamandé SR, and Shoulders MD
- Abstract
Objectives: Mutations in the procollagen-II gene ( COL2A1 ) often cause chondrodysplasias, including the precocious osteoarthritis-inducing p.Arg719Cys. Understanding the molecular basis of such diseases has long been challenging, owing to a lack of models accurately reflecting disease genotypes and phenotypes. To address this challenge, we develop and characterize in vitro human cartilage derived from wild-type and disease-causing Arg719Cys COL2A1 isogenic induced pluripotent stem cell (iPSC) lines., Methods: Using directed differentiation of iPSCs to chondrocytes, we generated cartilage from wild-type and Arg719Cys COL2A1 lines. We compared the resulting protein, cell, and tissue properties using immunohistochemistry, electron microscopy, SDS-PAGE, RNA-sequencing, and quantitative interactomics., Results: While both wild-type and disease lines deposited a cartilage matrix, the Arg719Cys matrix was deficient. Arg719Cys collagen-II was excessively post-translationally modified and modestly intracellularly retained, leading to endoplasmic reticulum (ER) distention suggestive of an ER storage defect. Interactomic studies indicated that Arg719Cys procollagen-II was not differentially engaged by the ER proteostasis network. RNA-sequencing showed that the ER storage defect engendered by Arg719Cys procollagen-II also did not activate cellular stress responses, including the unfolded protein response. These data suggest that cells fail to properly recognize Arg719Cys-associated procollagen-II defects., Conclusions: A failure to identify and rectify defective procollagen-II folding in cells expressing Arg719Cys procollagen-II leads to the deposition of a sparse and defective collagen-II matrix, culminating in pathology. Combined with the highly expandable human cartilage disease model reported here, this work provides motivation and a platform to discover therapeutic strategies targeting procollagen folding, quality control, and secretion in this collagenopathy and others.
- Published
- 2024
- Full Text
- View/download PDF
27. Dominant-negative TP53 mutations potentiated by the HSF1-regulated proteostasis network.
- Author
-
Sebastian RM, Patrick JE, Hui T, Amici DR, Giacomelli AO, Butty VL, Hahn WC, Mendillo ML, Lin YS, and Shoulders MD
- Abstract
Protein mutational landscapes are sculpted by the impacts of the resulting amino acid substitutions on the protein's stability and folding or aggregation kinetics. These properties can, in turn, be modulated by the composition and activities of the cellular proteostasis network. Heat shock factor 1 (HSF1) is the master regulator of the cytosolic and nuclear proteostasis networks, dynamically tuning the expression of cytosolic and nuclear chaperones and quality control factors to meet demand. Chronic increases in HSF1 levels and activity are prominent hallmarks of cancer cells. One plausible explanation for this observation is that the consequent upregulation of proteostasis factors could biophysically facilitate the acquisition of oncogenic mutations. Here, we experimentally evaluate the impacts of chronic HSF1 activation on the mutational landscape accessible to the quintessential oncoprotein p53. Specifically, we apply quantitative deep mutational scanning of p53 to assess how HSF1 activation shapes the mutational pathways by which p53 can escape cytotoxic pressure conferred by the small molecule nutlin-3, which is a potent antagonist of the p53 negative regulator MDM2. We find that activation of HSF1 broadly increases the fitness of dominant-negative substitutions within p53. This effect of HSF1 activation was particularly notable for non-conservative, biophysically unfavorable amino acid substitutions within buried regions of the p53 DNA-binding domain. These results indicate that chronic HSF1 activation profoundly shapes the oncogenic mutational landscape, preferentially supporting the acquisition of cancer-associated substitutions that are biophysically destabilizing. Along with providing the first experimental and quantitative insights into how HSF1 influences oncoprotein mutational spectra, these findings also implicate HSF1 inhibition as a strategy to reduce the accessibility of mutations that drive chemotherapeutic resistance and metastasis., Competing Interests: COMPETING INTERESTS W.C.H. is a consultant for Thermo Fischer Scientific, Solasta Ventures, KSQ Therapeutics, Frontier Medicines, Jubilant Therapeutics, RAPPTA Therapeutics, Serinus Biosciences, Kestral Therapeutics, Function Oncology, Crane Biotherapeutics and Perceptive. A.O.G. is a consultant for Atlas Venture.
- Published
- 2024
- Full Text
- View/download PDF
28. MutaT7 GDE : A Single Chimera for the Targeted, Balanced, Efficient, and Processive Installation of All Possible Transition Mutations In Vivo .
- Author
-
Mengiste AA, McDonald JL, Nguyen Tran MT, Plank AV, Wilson RH, Butty VL, and Shoulders MD
- Subjects
- Mutation, Mutagenesis, Directed Molecular Evolution methods, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Escherichia coli genetics, Viral Proteins genetics, Viral Proteins metabolism, DNA-Directed RNA Polymerases genetics, DNA-Directed RNA Polymerases metabolism
- Abstract
Deaminase-T7 RNA polymerase fusion (MutaT7) proteins are a growing class of synthetic biology tools used to diversify target genes during in vivo laboratory evolution. To date, MutaT7 chimeras comprise either a deoxyadenosine or deoxycytidine deaminase fused to a T7 RNA polymerase. Their expression drives targeted deoxyadenosine-to-deoxyguanosine or deoxycytidine-to-deoxythymidine mutagenesis, respectively. Here, we repurpose recently engineered substrate-promiscuous general deaminases (GDEs) to establish a substantially simplified system based on a single chimeric enzyme capable of targeting both deoxyadenosine and deoxycytidine. We assess on- and off-target mutagenesis, strand and context preference, and parity of deamination for four different MutaT7
GDE constructs. We identify a single chimera that installs all possible transition mutations more efficiently than preexisting, more cumbersome MutaT7 tools. The optimized MutaT7GDE chimera reported herein is a next-generation hypermutator capable of mediating efficient and uniform target-gene diversification during in vivo directed evolution.- Published
- 2024
- Full Text
- View/download PDF
29. ER procollagen storage defect without coupled unfolded protein response drives precocious arthritis.
- Author
-
Yammine KM, Mirda Abularach S, Kim SY, Bikovtseva AA, Lilianty J, Butty VL, Schiavoni RP, Bateman JF, Lamandé SR, and Shoulders MD
- Subjects
- Humans, Mutation, Induced Pluripotent Stem Cells metabolism, Cartilage metabolism, Cartilage pathology, Protein Folding, Arthritis metabolism, Arthritis genetics, Osteoarthritis metabolism, Osteoarthritis genetics, Osteoarthritis pathology, Animals, Chondrocytes metabolism, Unfolded Protein Response, Endoplasmic Reticulum metabolism, Procollagen metabolism, Collagen Type II metabolism
- Abstract
Collagenopathies are a group of clinically diverse disorders caused by defects in collagen folding and secretion. For example, mutations in the gene encoding collagen type-II, the primary collagen in cartilage, can lead to diverse chondrodysplasias. One example is the Gly1170Ser substitution in procollagen-II, which causes precocious osteoarthritis. Here, we biochemically and mechanistically characterize an induced pluripotent stem cell-based cartilage model of this disease, including both hetero- and homozygous genotypes. We show that Gly1170Ser procollagen-II is notably slow to fold and secrete. Instead, procollagen-II accumulates intracellularly, consistent with an endoplasmic reticulum (ER) storage disorder. Likely owing to the unique features of the collagen triple helix, this accumulation is not recognized by the unfolded protein response. Gly1170Ser procollagen-II interacts to a greater extent than wild-type with specific ER proteostasis network components, consistent with its slow folding. These findings provide mechanistic elucidation into the etiology of this disease. Moreover, the easily expandable cartilage model will enable rapid testing of therapeutic strategies to restore proteostasis in the collagenopathies., (© 2024 Yammine et al.)
- Published
- 2024
- Full Text
- View/download PDF
30. Emergent mechanical control of vascular morphogenesis.
- Author
-
Whisler J, Shahreza S, Schlegelmilch K, Ege N, Javanmardi Y, Malandrino A, Agrawal A, Fantin A, Serwinski B, Azizgolshani H, Park C, Shone V, Demuren OO, Del Rosario A, Butty VL, Holroyd N, Domart MC, Hooper S, Szita N, Boyer LA, Walker-Samuel S, Djordjevic B, Sheridan GK, Collinson L, Calvo F, Ruhrberg C, Sahai E, Kamm R, and Moeendarbary E
- Subjects
- Mice, Animals, Tissue Engineering methods, Morphogenesis, Cell Differentiation, Extracellular Matrix, Mechanotransduction, Cellular physiology, Endothelial Cells
- Abstract
Vascularization is driven by morphogen signals and mechanical cues that coordinately regulate cellular force generation, migration, and shape change to sculpt the developing vascular network. However, it remains unclear whether developing vasculature actively regulates its own mechanical properties to achieve effective vascularization. We engineered tissue constructs containing endothelial cells and fibroblasts to investigate the mechanics of vascularization. Tissue stiffness increases during vascular morphogenesis resulting from emergent interactions between endothelial cells, fibroblasts, and ECM and correlates with enhanced vascular function. Contractile cellular forces are key to emergent tissue stiffening and synergize with ECM mechanical properties to modulate the mechanics of vascularization. Emergent tissue stiffening and vascular function rely on mechanotransduction signaling within fibroblasts, mediated by YAP1. Mouse embryos lacking YAP1 in fibroblasts exhibit both reduced tissue stiffness and develop lethal vascular defects. Translating our findings through biology-inspired vascular tissue engineering approaches will have substantial implications in regenerative medicine.
- Published
- 2023
- Full Text
- View/download PDF
31. Ketone Body Signaling Mediates Intestinal Stem Cell Homeostasis and Adaptation to Diet.
- Author
-
Cheng CW, Biton M, Haber AL, Gunduz N, Eng G, Gaynor LT, Tripathi S, Calibasi-Kocal G, Rickelt S, Butty VL, Moreno-Serrano M, Iqbal AM, Bauer-Rowe KE, Imada S, Ulutas MS, Mylonas C, Whary MT, Levine SS, Basbinar Y, Hynes RO, Mino-Kenudson M, Deshpande V, Boyer LA, Fox JG, Terranova C, Rai K, Piwnica-Worms H, Mihaylova MM, Regev A, and Yilmaz ÖH
- Subjects
- 3-Hydroxybutyric Acid blood, 3-Hydroxybutyric Acid pharmacology, Aged, 80 and over, Animals, Cell Differentiation drug effects, Cell Self Renewal, Female, Histone Deacetylase Inhibitors pharmacology, Humans, Hydroxymethylglutaryl-CoA Synthase deficiency, Hydroxymethylglutaryl-CoA Synthase genetics, Hydroxymethylglutaryl-CoA Synthase metabolism, Intestines cytology, Intestines pathology, Male, Mice, Mice, Knockout, Receptors, G-Protein-Coupled metabolism, Receptors, Notch metabolism, Signal Transduction drug effects, Stem Cells cytology, Young Adult, Diet, High-Fat, Ketone Bodies metabolism, Stem Cells metabolism
- Abstract
Little is known about how metabolites couple tissue-specific stem cell function with physiology. Here we show that, in the mammalian small intestine, the expression of Hmgcs2 (3-hydroxy-3-methylglutaryl-CoA synthetase 2), the gene encoding the rate-limiting enzyme in the production of ketone bodies, including beta-hydroxybutyrate (βOHB), distinguishes self-renewing Lgr5
+ stem cells (ISCs) from differentiated cell types. Hmgcs2 loss depletes βOHB levels in Lgr5+ ISCs and skews their differentiation toward secretory cell fates, which can be rescued by exogenous βOHB and class I histone deacetylase (HDAC) inhibitor treatment. Mechanistically, βOHB acts by inhibiting HDACs to reinforce Notch signaling, instructing ISC self-renewal and lineage decisions. Notably, although a high-fat ketogenic diet elevates ISC function and post-injury regeneration through βOHB-mediated Notch signaling, a glucose-supplemented diet has the opposite effects. These findings reveal how control of βOHB-activated signaling in ISCs by diet helps to fine-tune stem cell adaptation in homeostasis and injury., (Copyright © 2019 Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
32. HRI coordinates translation necessary for protein homeostasis and mitochondrial function in erythropoiesis.
- Author
-
Zhang S, Macias-Garcia A, Ulirsch JC, Velazquez J, Butty VL, Levine SS, Sankaran VG, and Chen JJ
- Subjects
- Activating Transcription Factor 4 genetics, Anemia, Iron-Deficiency, Animals, Cell Differentiation, Erythroblasts, Eukaryotic Initiation Factor-2 metabolism, GRB10 Adaptor Protein genetics, GRB10 Adaptor Protein metabolism, Mice, Mice, Knockout, Oxygen metabolism, Phosphorylation, Protein Biosynthesis, Ribosomal Proteins, Unfolded Protein Response, eIF-2 Kinase genetics, Erythropoiesis physiology, Heme metabolism, Iron metabolism, Mitochondria metabolism, Proteostasis physiology, eIF-2 Kinase metabolism
- Abstract
Iron and heme play central roles in the production of red blood cells, but the underlying mechanisms remain incompletely understood. Heme-regulated eIF2α kinase (HRI) controls translation by phosphorylating eIF2α. Here, we investigate the global impact of iron, heme, and HRI on protein translation in vivo in murine primary erythroblasts using ribosome profiling. We validate the known role of HRI-mediated translational stimulation of integratedstressresponse mRNAs during iron deficiency in vivo. Moreover, we find that the translation of mRNAs encoding cytosolic and mitochondrial ribosomal proteins is substantially repressed by HRI during iron deficiency, causing a decrease in cytosolic and mitochondrial protein synthesis. The absence of HRI during iron deficiency elicits a prominent cytoplasmic unfolded protein response and impairs mitochondrial respiration. Importantly, ATF4 target genes are activated during iron deficiency to maintain mitochondrial function and to enable erythroid differentiation. We further identify GRB10 as a previously unappreciated regulator of terminal erythropoiesis., Competing Interests: SZ, AM, JU, JV, VB, SL, VS, JC No competing interests declared, (© 2019, Zhang et al.)
- Published
- 2019
- Full Text
- View/download PDF
33. Enhanced ER proteostasis and temperature differentially impact the mutational tolerance of influenza hemagglutinin.
- Author
-
Phillips AM, Doud MB, Gonzalez LO, Butty VL, Lin YS, Bloom JD, and Shoulders MD
- Subjects
- Amino Acid Sequence, Endoplasmic Reticulum Stress genetics, Gene Expression Profiling, HEK293 Cells, Hemagglutinin Glycoproteins, Influenza Virus genetics, Humans, Secretory Pathway genetics, Unfolded Protein Response genetics, Endoplasmic Reticulum metabolism, Hemagglutinin Glycoproteins, Influenza Virus metabolism, Mutation, Proteostasis, Temperature
- Abstract
We systematically and quantitatively evaluate whether endoplasmic reticulum (ER) proteostasis factors impact the mutational tolerance of secretory pathway proteins. We focus on influenza hemaggluttinin (HA), a viral membrane protein that folds in the host's ER via a complex pathway. By integrating chemical methods to modulate ER proteostasis with deep mutational scanning to assess mutational tolerance, we discover that upregulation of ER proteostasis factors broadly enhances HA mutational tolerance across diverse structural elements. Remarkably, this proteostasis network-enhanced mutational tolerance occurs at the same sites where mutational tolerance is most reduced by propagation at fever-like temperature. These findings have important implications for influenza evolution, because influenza immune escape is contingent on HA possessing sufficient mutational tolerance to evade antibodies while maintaining the capacity to fold and function. More broadly, this work provides the first experimental evidence that ER proteostasis mechanisms define the mutational tolerance and, therefore, the evolution of secretory pathway proteins., Competing Interests: AP, MD, LG, VB, YL, JB, MS No competing interests declared, (© 2018, Phillips et al.)
- Published
- 2018
- Full Text
- View/download PDF
34. Anti-inflammatory response after infusion of p55 soluble tumor necrosis factor receptor fusion protein for severe sepsis.
- Author
-
Butty VL, Roux-Lombard P, Garbino J, Dayer JM, and Ricou B
- Subjects
- Double-Blind Method, Female, Humans, Immunoglobulin gamma-Chains, Intensive Care Units, Male, Middle Aged, Placebos, Sepsis blood, Sepsis immunology, Shock, Septic blood, Shock, Septic immunology, Treatment Outcome, Cytokines blood, Immunoglobulin G therapeutic use, Immunoglobulin Heavy Chains, Receptors, Tumor Necrosis Factor therapeutic use, Recombinant Fusion Proteins therapeutic use, Sepsis drug therapy, Shock, Septic drug therapy, Tumor Necrosis Factor-alpha antagonists & inhibitors
- Abstract
Objectives: To investigate the effects of Lenercept , a recombinant soluble TNF receptor p55 fused to an immunoglobulin heavy chain IgG1, on the balance of pro- and anti-inflammatory mediators in sepsis., Design: Post hoc analysis of a subgroup of patients enrolled in a multicenter phase III, prospective, double-blind, placebo-controlled, randomized study of Lenercept in severe sepsis., Setting: Surgical and medical intensive care units, and postoperative recovery room of a tertiary care teaching hospital., Patients: A total of 57 patients were enrolled in the multicenter study in our center., Intervention: Septic patients were randomly assigned to receive either Lenercept 0.125 mg/kg or placebo. The patients were followed for up to 28 days after randomization., Measurements and Main Results: Circulating levels of TNF-alpha, IL-6, TNFsR75 and IL-1Ra were measured before and after treatment. The two groups were comparable with regard to age, gender and diagnosis distribution. The total level of TNF-alpha increased significantly in treated patients, compared to patients receiving placebo. The levels of the other inflammatory mediators did not differ between the two groups, Conclusions: Lenercept -treated patients experienced a protracted TNF-alpha half-life, leading to higher total TNF-alpha levels throughout the study. However, the treatment had no effects on anti-inflammatory mediators. Therefore, peripheral inflammatory processes might not have been significantly modified by the treatment. This might account for the lack of efficacy this treatment in septic patients
- Published
- 2003
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.