Network


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

Hotspot


Dive into the research topics where David J. Busch is active.

Publication


Featured researches published by David J. Busch.


Nature Communications | 2015

Intrinsically disordered proteins drive membrane curvature

David J. Busch; Justin R. Houser; Carl C. Hayden; Michael B. Sherman; Eileen M. Lafer; Jeanne C. Stachowiak

Assembly of highly curved membrane structures is essential to cellular physiology. The prevailing view has been that proteins with curvature-promoting structural motifs, such as wedge-like amphipathic helices and crescent-shaped BAR domains, are required for bending membranes. Here we report that intrinsically disordered domains of the endocytic adaptor proteins, Epsin1 and AP180 are highly potent drivers of membrane curvature. This result is unexpected since intrinsically disordered domains lack a well-defined three-dimensional structure. However, in vitro measurements of membrane curvature and protein diffusivity demonstrate that the large hydrodynamic radii of these domains generate steric pressure that drives membrane bending. When disordered adaptor domains are expressed as transmembrane cargo in mammalian cells, they are excluded from clathrin-coated pits. We propose that a balance of steric pressure on the two surfaces of the membrane drives this exclusion. These results provide quantitative evidence for the influence of steric pressure on the content and assembly of curved cellular membrane structures.


The Journal of Comparative Neurology | 2012

Synuclein accumulation is associated with cell‐specific neuronal death after spinal cord injury

David J. Busch; Jennifer R. Morgan

Spinal cord injury axotomizes neurons and induces many of them to die, whereas others survive. Therefore, it is important to identify factors that lead to neuronal death after injury as a first step toward developing better strategies for increasing neuronal survival and functional recovery. However, the intrinsic molecular pathways that govern whether an injured neuron lives or dies remain surprisingly unclear. To address this question, we took advantage of the large size of giant reticulospinal (RS) neurons in the brain of the lamprey, Petromyzon marinus. We report that axotomy of giant RS neurons induces a select subset of them to accumulate high levels of synuclein, a synaptic vesicle‐associated protein whose abnormal accumulation is linked to Parkinsons disease. Injury‐induced synuclein accumulation occurred only in neurons that were classified as “poor survivors” by both histological and Fluoro‐Jade C staining. In contrast, post‐injury synuclein immunofluorescence remained at control levels in neurons that were identified as “good survivors.” Synuclein accumulation appeared in the form of aggregated intracellular inclusions. Cells that accumulated synuclein also exhibited more ubiquitin‐containing inclusions, similar to what occurs during disease states. When synuclein levels and cell vitality were measured in the same neurons, it became clear that synuclein accumulation preceded and strongly correlated with subsequent neuronal death. Thus, synuclein accumulation is identified as a marker and potential risk factor for forthcoming neuronal death after axotomy, expanding its implications beyond the neurodegenerative diseases. J. Comp. Neurol. 520:1751–1771, 2012.


Molecular Biology of the Cell | 2014

Acute increase of α-synuclein inhibits synaptic vesicle recycling evoked during intense stimulation.

David J. Busch; Paul A. Oliphint; Rylie B. Walsh; Susan M. L. Banks; Wendy S. Woods; Julia M. George; Jennifer R. Morgan

This is the first study to show the direct effects of α-synuclein on synaptic vesicle trafficking and to elucidate the underlying structural mechanisms. Acutely increasing α-synuclein severely inhibits synaptic vesicle recycling from the plasma membrane. The endocytic defects require a properly folded N-terminal α-helical domain of α-synuclein.


Experimental Neurology | 2011

Increased synapsin expression and neurite sprouting in lamprey brain after spinal cord injury

Billy Y.B. Lau; Andrea E. Foldes; Naila O. Alieva; Paul A. Oliphint; David J. Busch; Jennifer R. Morgan

Spinal cord injury induces structural plasticity throughout the mammalian nervous system, including distant locations in the brain. Several types of injury-induced plasticity have been identified, such as neurite sprouting, axon regeneration, and synaptic remodeling. However, the molecular mechanisms involved in injury-induced plasticity are unclear as is the extent to which injury-induced plasticity in brain is conserved across vertebrate lineages. Due to its robust roles in neurite outgrowth and synapse formation during developmental processes, we examined synapsin for its potential involvement in injury-induced plasticity. We used lamprey, a vertebrate that undergoes robust anatomical plasticity and functional recovery after spinal cord injury. At 3 and 11 weeks after spinal cord transection, synapsin I mRNA was upregulated >2-fold in lamprey brain, as assayed by semi-quantitative RT-PCR. Other synaptic vesicle-associated genes remained unchanged. In situ hybridization revealed that synapsin I mRNA was increased globally throughout the lamprey brain. Immunolabeling for synapsin I protein revealed a significant increase in both the intensity and density of synapsin I-positive structures in lamprey hindbrain at 11 weeks post-transection, relative to controls. Moreover, the number of structures immunolabeled for phospho-synapsin (serine 9) increased after injury, suggestive of neurite sprouting. Indeed, at the ultrastructural level, there was an increase in neurite density at 11 weeks post-transection. Taken together, these data show that neurite sprouting in the brain is an evolutionarily conserved response to a distant spinal cord injury and suggest that synapsin and its phosphorylation at serine 9 play key roles in the sprouting mechanism.


The Journal of Neuroscience | 2013

A Role for an Hsp70 Nucleotide Exchange Factor in the Regulation of Synaptic Vesicle Endocytosis

Jennifer R. Morgan; Jianwen Jiang; Paul A. Oliphint; Suping Jin; Luis E. Gimenez; David J. Busch; Andrea E. Foldes; Yue Zhuo; Rui Sousa; Eileen M. Lafer

Neurotransmission requires a continuously available pool of synaptic vesicles (SVs) that can fuse with the plasma membrane and release their neurotransmitter contents upon stimulation. After fusion, SV membranes and membrane proteins are retrieved from the presynaptic plasma membrane by clathrin-mediated endocytosis. After the internalization of a clathrin-coated vesicle, the vesicle must uncoat to replenish the pool of SVs. Clathrin-coated vesicle uncoating requires ATP and is mediated by the ubiquitous molecular chaperone Hsc70. In vitro, depolymerized clathrin forms a stable complex with Hsc70*ADP. This complex can be dissociated by nucleotide exchange factors (NEFs) that release ADP from Hsc70, allowing ATP to bind and induce disruption of the clathrin:Hsc70 association. Whether NEFs generally play similar roles in vesicle trafficking in vivo and whether they play such roles in SV endocytosis in particular is unknown. To address this question, we used information from recent structural and mechanistic studies of Hsp70:NEF and Hsp70:co-chaperone interactions to design a NEF inhibitor. Using acute perturbations at giant reticulospinal synapses of the sea lamprey (Petromyzon marinus), we found that this NEF inhibitor inhibited SV endocytosis. When this inhibitor was mutated so that it could no longer bind and inhibit Hsp110 (a NEF that we find to be highly abundant in brain cytosol), its ability to inhibit SV endocytosis was eliminated. These observations indicate that the action of a NEF, most likely Hsp110, is normally required during SV trafficking to release clathrin from Hsc70 and make it available for additional rounds of endocytosis.


Small | 2016

Multifunctional Transmembrane Protein Ligands for Cell-Specific Targeting of Plasma Membrane-Derived Vesicles.

Chi Zhao; David J. Busch; Connor P. Vershel; Jeanne C. Stachowiak

Liposomes and nanoparticles that bind selectively to cell-surface receptors can target specific populations of cells. However, chemical conjugation of ligands to these particles is difficult to control, frequently limiting ligand uniformity and complexity. In contrast, the surfaces of living cells are decorated with highly uniform populations of sophisticated transmembrane proteins. Toward harnessing cellular capabilities, here it is demonstrated that plasma membrane vesicles (PMVs) derived from donor cells can display engineered transmembrane protein ligands that precisely target cells on the basis of receptor expression. These multifunctional targeting proteins incorporate (i) a protein ligand, (ii) an intrinsically disordered protein spacer to make the ligand sterically accessible, and (iii) a fluorescent protein domain that enables quantification of the ligand density on the PMV surface. PMVs that display targeting proteins with affinity for the epidermal growth factor receptor (EGFR) bind at increasing concentrations to breast cancer cells that express increasing levels of EGFR. Further, as an example of the generality of this approach, PMVs expressing a single-domain antibody against green fluorescence protein (eGFP) bind to cells expressing eGFP-tagged receptors with a selectivity of ≈50:1. The results demonstrate the versatility of PMVs as cell targeting systems, suggesting diverse applications from drug delivery to tissue engineering.


Biophysical Journal | 2018

Entropic Control of Receptor Recycling Using Engineered Ligands

Andre C.M. DeGroot; David J. Busch; Carl C. Hayden; Samuel A. Mihelic; Aaron T. Alpar; Marcelo Behar; Jeanne C. Stachowiak

Receptor internalization by endocytosis regulates diverse cellular processes, from the rate of nutrient uptake to the timescale of essential signaling events. The established view is that internalization is tightly controlled by specific protein-binding interactions. However, recent work suggests that physical aspects of receptors influence the process in ways that cannot be explained by biochemistry alone. Specifically, work from several groups suggests that increasing the steric bulk of receptors may inhibit their uptake by multiple types of trafficking vesicles. How do biochemical and biophysical factors work together to control internalization? Here, we show that receptor uptake is well described by a thermodynamic trade-off between receptor-vesicle binding energy and the entropic cost of confining receptors within endocytic vesicles. Specifically, using large ligands to acutely increase the size of engineered variants of the transferrin receptor, we demonstrate that an increase in the steric bulk of a receptor dramatically decreases its probability of uptake by clathrin-coated structures. Further, in agreement with a simple thermodynamic analysis, all data collapse onto a single trend relating fractional occupancy of the endocytic structure to fractional occupancy of the surrounding plasma membrane, independent of receptor size. This fundamental scaling law provides a simple tool for predicting the impact of receptor expression level, steric bulk, and the size of endocytic structures on receptor uptake. More broadly, this work suggests that bulky ligands could be used to drive the accumulation of specific receptors at the plasma membrane surface, providing a biophysical tool for targeted modulation of signaling and metabolism from outside the cell.


Small | 2016

Biomaterials: Multifunctional Transmembrane Protein Ligands for Cell‐Specific Targeting of Plasma Membrane‐Derived Vesicles (Small 28/2016)

Chi Zhao; David J. Busch; Connor P. Vershel; Jeanne C. Stachowiak

Cell-derived plasma membrane vesicles extracted from genetically engineered donor cells can selectively target cells based on their receptor expression profiles, as demonstrated by J. C. Stachowiak and co-workers on page 3837. These engineered donor cells have a stable expression of transmembrane protein ligands that contain multiple domains, including a targeting ligand, a spacer and a fluorophore.


Soft Matter | 2015

Designing lipids for selective partitioning into liquid ordered membrane domains

Noor Momin; Stacey Lee; Avinash K. Gadok; David J. Busch; George D. Bachand; Carl C. Hayden; Jeanne C. Stachowiak; Darryl Y. Sasaki


Biophysical Journal | 2016

Connectosomes for Direct Molecular Delivery to the Cellular Cytoplasm

Avinash K. Gadok; David J. Busch; Silvia Ferrati; Brian Li; Hugh D. C. Smyth; Jeanne C. Stachowiak

Collaboration


Dive into the David J. Busch's collaboration.

Top Co-Authors

Avatar

Jeanne C. Stachowiak

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Carl C. Hayden

Sandia National Laboratories

View shared research outputs
Top Co-Authors

Avatar

Jennifer R. Morgan

Marine Biological Laboratory

View shared research outputs
Top Co-Authors

Avatar

Aaron T. Alpar

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Chi Zhao

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Justin R. Houser

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Paul A. Oliphint

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Andre C.M. DeGroot

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Andrea E. Foldes

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Avinash K. Gadok

University of Texas at Austin

View shared research outputs
Researchain Logo
Decentralizing Knowledge