Global Highlights in Neuroengineering 2005-2018


1 Comment

PDF version: 

Global Highlights in Neuroengineering 2005-2018 – Logan Thrasher Collins

Optogenetic stimulation using ChR2

(Boyden, Zhang, Bamberg, Nagel, & Deisseroth, 2005)

  • Ed S. Boyden, Karl Deisseroth, and colleagues developed optogenetics, a revolutionary technique for stimulating neural activity.
  • Optogenetics involves engineering neurons to express light-gated ion channels. The first channel used for this purpose was ChR2 (a protein originally found in bacteria which responds to blue light). In this way, a neuron exposed to an appropriate wavelength of light will be stimulated.
  • Over time, optogenetics has gained a place as an essential experimental tool for neuroscientists across the world. It has been expanded upon and improved in numerous ways and has even allowed control of animal behavior via implanted fiber optics and other light sources. Optogenetics may eventually be used in the development of improved brain-computer interfaces.

Optogenetics

Blue Brain Project cortical column simulation

(Markram, 2006)

  • In the early stages of the Blue Brain Project, ~ 10,000 neurons were mapped in 2-week-old rat somatosensory neocortical columns with sufficient resolution to show rough spatial locations of the dendrites and synapses.
  • After constructing a virtual model, algorithmic adjustments refined the spatial connections between neurons to increase accuracy (over 10 million synapses).
  • The cortical column was emulated using the Blue Gene/L supercomputer and the emulation was highly accurate compared to experimental data.

Cortical Column

Optogenetic silencing using halorhodopsin

(Han & Boyden, 2007)

  • Ed Boyden continued developing optogenetic tools to manipulate neural activity. Along with Xue Han, he expressed a codon-optimized version of a bacterial halorhodopsin (along with the ChR2 protein) in neurons.
  • Upon exposure to yellow light, halorhodopsin pumps chloride ions into the cell, hyperpolarizing the membrane and inhibiting neural activity.
  • Using halorhodopsin and ChR2, neurons could be easily activated and inhibited using yellow and blue light respectively.

Halorhodopsin and ChR2 wavelengths

Brainbow

(Livet et al., 2007)

  • Lichtman and colleagues used Cre/Lox recombination tools to create genes which express a randomized set of three or more differently-colored fluorescent proteins (XFPs) in a given neuron, labeling the neuron with a unique combination of colors. About ninety distinct colors were emitted across a population of genetically modified neurons.
  • The detailed structures within neural tissue equipped with the Brainbow system can be imaged much more easily since neurons can be distinguished via color contrast.
  • As a proof-of-concept, hundreds of synaptic contacts and axonal processes were reconstructed in a selected volume of the cerebellum. Several other neural structures were also imaged using Brainbow.
  • The fluorescent proteins expressed by the Brainbow system are usable in vivo.

Brainbow

High temporal precision optogenetics

(Gunaydin et al., 2010)

  • Karl Deisseroth, Peter Hegemann, and colleagues used protein engineering to improve the temporal resolution of optogenetic stimulation.
  • Glutamic acid at position 123 in ChR2 was mutated to threonine, producing a new ion channel protein (dubbed ChETA).
  • The ChETA protein allows for induction of spike trains with frequencies up to 200 Hz and greatly decreases the incidence of unintended spikes. Furthermore, ChETA eliminates plateau potentials (a phenomenon which interferes with precise control of neural activity).

Ultrafast optogenetics

Hippocampal prosthesis in rats

(Berger et al., 2012)

  • Theodore Berger and his team developed an artificial replacement for neurons which transmit information from the CA3 region to the CA1 region of the hippocampus.
  • This cognitive prosthesis employs recording and stimulation electrodes along with a multi-input multi-output (MIMO) model to encode the information in CA3 and transfer it to CA1.
  • The hippocampal prosthesis was shown to restore and enhance memory in rats as evaluated by behavioral testing and brain imaging.

In vivo superresolution microscopy for neuroimaging

(Berning, Willig, Steffens, Dibaj, & Hell, 2012)

  • Stefan Hell (2014 Nobel laureate in chemistry) developed stimulated emission depletion microscopy (STED), a type of superresolution fluorescence microscopy which allows imaging of synapses and dendritic spines.
  • STED microscopy uses transgenic neurons that express fluorescent proteins. The neurons exhibit fluctuations in their fluorescence over time, providing temporal contrast enhancement to the resolution. Although light’s wavelength would ordinarily limit the resolution (the diffraction limit), STED’s temporal contrast overcomes this limitation.
  • Neurons in transgenic mice (equipped with glass-sealed holes in their skulls) were imaged using STED. Synapses and dendritic spines were observed up to fifteen nanometers below the surface of the brain tissue.

Superresolution microscopy in vivo

Eyewire: crowdsourcing method for retina mapping

(Marx, 2013)

  • The Eyewire project was created by Sebastian Seung’s research group. It is a crowdsourcing initiative for connectomic mapping within the retina towards uncovering neural circuits involved in visual processing.
  • Laboratories first collect data via serial electron microscopy as well as functional data from two-photon microscopy.
  • In the Eyewire game, images of tissue slices are provided to players who then help reconstruct neural morphologies and circuits by “coloring in” the parts of the images which correspond to cells and stacking many images on top of each other to generate 3D maps. Artificial intelligence tools help provide initial “best guesses” and guide the players, but the people ultimately perform the task of reconstruction.
  • By November 2013, around 82,000 participants had played the game. Its popularity continues to grow.

Eyewire

The BRAIN Initiative

(“Fact Sheet: BRAIN Initiative,” 2013)

  • The BRAIN Initiative (Brain Research through Advancing Innovative Technologies) provided neuroscientists with $110 million in governmental funding and $122 million in funding from private sources such as the Howard Hughes Medical Institute and the Allen Institute for Brain Science.
  • The BRAIN Initiative focused on funding research which develops and utilizes new technologies for functional connectomics. It helped to accelerate research on tools for decoding the mechanisms of neural circuits in order to understand and treat mental illness, neurodegenerative diseases, and traumatic brain injury.
  • The BRAIN Initiative emphasized collaboration between neuroscientists and physicists. It also pushed forward nanotechnology-based methods to image neural tissue, record from neurons, and otherwise collect neurobiological data.

The CLARITY method for making brains translucent

(Chung & Deisseroth, 2013)

  • Karl Deisseroth and colleagues developed a method called CLARITY to make samples of neural tissue optically translucent without damaging the fine cellular structures in the tissue. Using CLARITY, entire mouse brains have been turned transparent.
  • Mouse brains were infused with hydrogel monomers (acrylamide and bisacrylamide) as well as formaldehyde and some other compounds for facilitating crosslinking. Next, the hydrogel monomers were crosslinked by incubating the brains at 37°C. Lipids in the hydrogel-stabilized mouse brains were extracted using hydrophobic organic solvents and electrophoresis.
  • CLARITY allows antibody labeling, fluorescence microscopy, and other optically-dependent techniques to be used for imaging entire brains. In addition, it renders the tissue permeable to macromolecules, which broadens the types of experimental techniques that these samples can undergo (i.e. macromolecule-based stains, etc.)

CLARITY Imaging TechniqueTelepathic rats engineered using hippocampal prosthesis

(S. Deadwyler et al., 2013)

  • Berger’s hippocampal prosthesis was implanted in pairs of rats. When “donor” rats were trained to perform a task, they developed neural representations (memories) which were recorded by their hippocampal prostheses.
  • The donor rat memories were run through the MIMO model and transmitted to the stimulation electrodes of the hippocampal prostheses implanted in untrained “recipient” rats. After receiving the memories, the recipient rats showed significant improvements on the task that they had not been trained to perform.

Rat Telepathy

Integrated Information Theory 3.0

(Oizumi, Albantakis, & Tononi, 2014)

  • Integrated information theory (IIT) was originally proposed by Giulio Tononi in 2004. IIT is a quantitative theory of consciousness which may help explain the hard problem of consciousness.
  • IIT begins by assuming the following phenomenological axioms; each experience is characterized by how it differs from other experiences, an experience cannot be reduced to interdependent parts, and the boundaries which distinguish individual experiences are describable as having defined “spatiotemporal grains.”
  • From these phenomenological axioms and the assumption of causality, IIT identifies maximally irreducible conceptual structures (MICS) associated with individual experiences. MICS represent particular patterns of qualia that form unified percepts.
  • IIT also outlines a mathematical measure of an experience’s quantity. This measure is called integrated information or ϕ.

Expansion Microscopy

(F. Chen, Tillberg, & Boyden, 2015)

  • The Boyden group developed expansion microscopy, a method which enlarges neural tissue samples (including entire brains) with minimal structural distortions and so facilitates superior optical visualization of the scaled-up neural microanatomy. Furthermore, expansion microscopy greatly increases the optical translucency of treated samples.
  • Expansion microscopy operates by infusing a swellable polymer network into brain tissue samples along with several chemical treatments to facilitate polymerization and crosslinking and then triggering expansion via dialysis in water. With 4.5-fold enlargement, expansion microscopy only distorts the tissue by about 1% (computed using a comparison between control superresolution microscopy of easily-resolvable cellular features and the expanded version).
  • Before expansion, samples can express various fluorescent proteins to facilitate superresolution microscopy of the enlarged tissue once the process is complete. Furthermore, expanded tissue is highly amenable to fluorescent stains and antibody-based labels.

Expansion microscopy

Japan’s Brain/MINDS project

(Okano, Miyawaki, & Kasai, 2015)

  • In 2014, the Brain/MINDS (Brain Mapping by Integrated Neurotechnologies for Disease Studies) project was initiated to further neuroscientific understanding of the brain. This project received nearly $30 million in funding for its first year alone.
  • Brain/MINDS focuses on studying the brain of the common marmoset (a non-human primate abundant in Japan), developing new technologies for brain mapping, and understanding the human brain with the goal of finding new treatments for brain diseases.

Openworm

(Szigeti et al., 2014)

  • The anatomical elegans connectome was originally mapped in 1976 by Albertson and Thomson. More data has since been collected on neurotransmitters, electrophysiology, cell morphology, and other characteristics.
  • Szigeti, Larson, and their colleagues made an online platform for crowdsourcing research on elegans computational neuroscience, with the goal of completing an entire “simulated worm.”
  • The group also released software called Geppetto, a program that allows users to manipulate both multicompartmental Hodgkin-Huxley models and highly efficient soft-body physics simulations (for modeling the worm’s electrophysiology and anatomy).

C. elegans Connectome

The TrueNorth Chip from DARPA and IBM

(Akopyan et al., 2015)

  • The TrueNorth neuromorphic computing chip was constructed and validated by DARPA and IBM. TrueNorth uses circuit modules which mimic neurons. Inputs to these fundamental circuit modules must overcome a threshold in order to trigger “firing.”
  • The chip can emulate up to a million neurons with over 250 million synapses while requiring far less power than traditional computing devices.

Human Brain Project cortical mesocircuit reconstruction and simulation

(Markram et al., 2015)

  • The HBP digitally reconstructed a 0.29 mm3 region of rat cortical tissue (~ 31,000 neurons and 37 million synapses) based on morphological data, “connectivity rules,” and additional datasets. The cortical mesocircuit was emulated using the Blue Gene/Q supercomputer.
  • This emulation was sufficiently accurate to reproduce emergent neurological processes and yield insights on the mechanisms of their computations.

Cortical Mesocircuit

Neural lace

(Liu et al., 2015)

  • Charles Lieber’s group developed a syringe-injectable electronic mesh made of submicrometer-thick wiring for neural interfacing.
  • The meshes were constructed using novel soft electronics for biocompatibility. Upon injection, the neural lace expands to cover and record from centimeter-scale regions of tissue.
  • Neural lace may allow for “invasive” brain-computer interfaces to circumvent the need for surgical implantation. Lieber has continued to develop this technology towards clinical application.

Neural Lace

Expansion FISH

(F. Chen et al., 2016)

  • Boyden, Chen, Marblestone, Church, and colleagues combined fluorescent in situ hybridization (FISH) with expansion microscopy to image the spatial localization of RNA in neural tissue.
  • The group developed a chemical linker to covalently attach intracellular RNA to the infused polymer network used in expansion microscopy. This allowed for RNAs to maintain their relative spatial locations within each cell post-expansion.
  • After the tissue was enlarged, FISH was used to fluorescently label targeted RNA molecules. In this way, RNA localization was more effectively resolved.
  • As a proof-of-concept, expansion FISH was used to reveal the nanoscale distribution of long noncoding RNAs in nuclei as well as the locations of RNAs within dendritic spines.

Expansion FISH

Neural dust

(Seo et al., 2016)

  • Michel Maharbiz’s group invented implantable, ~ 1 mm biosensors for wireless neural recording and tested them in rats.
  • This neural dust could be miniaturized to less than 0.5 mm or even to microscale dimensions using customized electronic components.
  • Neural dust motes consist of two recording electrodes, a transistor, and a piezoelectric crystal.
  • The neural dust received external power from ultrasound. Neural signals were recorded by measuring disruptions to the piezoelectric crystal’s reflection of the ultrasound waves. Signal processing mathematics allowed precise detection of activity.

Neural Dust

The China Brain Project

(Poo et al., 2016)

  • The China Brain Project was launched to help understand the neural mechanisms of cognition, develop brain research technology platforms, develop preventative and diagnostic interventions for brain disorders, and to improve brain-inspired artificial intelligence technologies.
  • This project will be take place from 2016 until 2030 with the goal of completing mesoscopic brain circuit maps.
  • China’s population of non-human primates and preexisting non-human primate research facilities give the China Brain Project an advantage. The project will focus on studying rhesus macaques.

Somatosensory cortex stimulation for spinal cord injuries

(Flesher et al., 2016)

  • Gaunt, Flesher, and colleagues found that microstimulation of the primary somatosensory cortex (S1) partially restored tactile sensations to a patient with a spinal cord injury.
  • Electrode arrays were implanted into the S1 regions of a patient with a spinal cord injury. The array performed intracortical microstimulation over a period of six months.
  • The patient reported locations and perceptual qualities of the sensations elicited by microstimulation. The patient did not experience pain or “pins and needles” from any of the stimulus trains. Overall, 93% of the stimulus trains were reported as “possibly natural.”
  • Results from this study might be used to engineer upper-limb neuroprostheses which provide somatosensory feedback.

Somatosensory Stimulation

Hippocampal prosthesis in monkeys

(S. A. Deadwyler et al., 2017)

  • Theodore Berger continued developing his cognitive prosthesis and tested it in Rhesus Macaques.
  • As with the rats, monkeys with the implant showed substantially improved performance on memory tasks.

The $100 billion Softbank Vision Fund

(Lomas, 2017)

  • Masayoshi Son, the CEO of Softbank (a Japanese telecommunications corporation), announced a plan to raise $100 billion in venture capital to invest in artificial intelligence. This plan involved partnering with multiple large companies in order to raise this enormous amount of capital.
  • By the end of 2017, the Vision Fund successfully reached its $100 billion goal. Masayoshi Son has since announced further plans to continue raising money with a new goal of over $800 billion.
  • Masayoshi Son’s reason for these massive investments is the Technological Singularity. He agrees with Kurzweil that the Singularity will likely occur at around 2045 and he hopes to help bring the Singularity to fruition. Though Son is aware of the risks posed by artificial superintelligence, he feels that superintelligent AI’s potential to tackle some of humanity’s greatest challenges (such as climate change and the threat of nuclear war) outweighs those risks.

Bryan Johnson launches Kernel

(Regalado, 2017)

  • Entrepreneur Bryan Johnson invested $100 million to start Kernel, a neurotechnology company.
  • Kernel plans to develop implants that allow for recording and stimulation of large numbers of neurons at once. The company’s initial goal is to develop treatments for mental illnesses and neurodegenerative diseases. Its long-term goal is to enhance human intelligence.
  • Kernel originally partnered with Theodore Berger and intended to utilize his hippocampal prosthesis. Unfortunately, Berger and Kernel parted ways after about six months because Berger’s vision was reportedly too long-range to support a financially viable company (at least for now).
  • Kernel was originally a company called Kendall Research Systems. This company was started by a former member of the Boyden lab. In total, four members of Kernel’s team are former Boyden lab members.

Elon Musk launches NeuraLink

(Etherington, 2017)

  • Elon Musk (CEO of Tesla, SpaceX, and a number of other successful companies) initiated a neuroengineering venture called NeuraLink.
  • NeuraLink will begin by developing brain-computer interfaces (BCIs) for clinical applications, but the ultimate goal of the company is to enhance human cognitive abilities in order to keep up with artificial intelligence.
  • Though many of the details around NeuraLink’s research are not yet open to the public, it has been rumored that injectable electronics similar to Lieber’s neural lace might be involved.

Facebook announces effort to build brain-computer interfaces

(Constine, 2017)

  • Facebook revealed research on constructing non-invasive brain-computer interfaces (BCIs) at a company-run conference in 2017. The initiative is run by Regina Dugan, Facebook’s head of R&D at division building 8.
  • Facebook’s researchers are working on a non-invasive BCI which may eventually enable users to type one hundred words per minute with their thoughts alone. This effort builds on past investigations which have been used to help paralyzed patients.
  • The building 8 group is also developing a wearable device for “skin hearing.” Using just a series of vibrating actuators which mimic the cochlea, test subjects have so far been able to recognize up to nine words. Facebook intends to vastly expand this device’s capabilities.

DARPA funds research to develop improved brain-computer interfaces

(Hatmaker, 2017)

  • The U.S. government agency DARPA awarded $65 million in total funding to six research groups.
  • The recipients of this grant included five academic laboratories (headed by Arto Nurmikko, Ken Shepard, Jose-Alain Sahel and Serge Picaud, Vicent Pieribone, and Ehud Isacoff) and one small company called Paradromics Inc.
  • DARPA’s goal for this initiative is to develop a nickel-sized bidirectional brain-computer interface (BCI) which can record from and stimulate up to one million individual neurons at once.

Human Brain Project analyzes brain computations using algebraic topology

(Reimann et al., 2017)

  • Investigators at the Human Brain Project utilized algebraic topology to analyze the reconstructed ~ 31,000 neuron cortical microcircuit from their earlier work.
  • The analysis involved representing the cortical network as a digraph, finding directed cliques (complete directed subgraphs belonging to a digraph), and determining the net directionality of information flow (by computing the sum of the squares of the differences between in-degree and out-degree for all the neurons in a clique). In algebraic topology, directed cliques of n neurons are called directed simplices of dimension n-1.
  • Vast numbers of high-dimensional directed cliques were found in the cortical microcircuit (as compared to null models and other controls). Spike correlations between pairs of neurons within a clique were found to increase with the clique’s dimension and with the proximity of the neurons to the clique’s sink. Furthermore, topological metrics allowed insights into the flow of neural information among multiple cliques.
  • Experimental patch-clamp data supported the significance of the findings. In addition, similar patterns were found within the C. elegans connectome, suggesting that the results may generalize to nervous systems across species.

HBP algebraic topology

Early testing of hippocampal prosthesis algorithm in humans

(Song, She, Hampson, Deadwyler, & Berger, 2017)

  • Dong Song (who was working alongside Berger) tested the MIMO algorithm on human epilepsy patients using implanted recording and stimulation electrodes. The full hippocampal prosthesis was not implanted, but the electrodes acted similarly, though in a temporary capacity. Although only two patients were tested in this study, many trials were performed to compensate for the small sample size.
  • Hippocampal spike trains from individual cells in CA1 and CA3 were recorded from the patients during a delayed match-to-sample task. The patients were shown various images while neural activity data were recorded by the electrodes and processed by the MIMO model. The patients were then asked to recall which image they had been shown previously by picking it from a group of “distractor” images. Memories encoded by the MIMO model were used to stimulate hippocampal cells during the recall phase.
  • In comparison to controls in which the same two epilepsy patients were not assisted by the algorithm and stimulation, the experimental trials demonstrated a significant increase in successful pattern matching.

Brain imaging factory in China

(Cyranoski, 2017)

  • Qingming Luo started the HUST-Suzhou Institute for Brainsmatics, a brain imaging “factory.” Each of the numerous machines in Luo’s facility performs automated processing and imaging of tissue samples. The devices make ultrathin slices of brain tissue using diamond blades, treat the samples with fluorescent stains or other contrast-enhancing chemicals, and image then using fluorescence microscopy.
  • The institute has already demonstrated its potential by mapping the morphology of a previously unknown neuron which “wraps around” the entire mouse brain.

China Brain Mapping Image

Automated patch-clamp robot for in vivo neural recording

(Suk et al., 2017)

  • Ed S. Boyden and colleagues developed a robotic system to automate patch-clamp recordings from individual neurons. The robot was tested in vivo using mice and achieved a data collection yield similar to that of skilled human experimenters.
  • By continuously imaging neural tissue using two-photon microscopy, the robot can adapt to a target cell’s movement and shift the pipette to compensate. This adaptation is facilitated by a novel algorithm called an imagepatching algorithm. As the pipette approaches its target, the algorithm adjusts the pipette’s trajectory based on the real-time two-photon microscopy.
  • The robot can be used in vivo so long as the target cells express a fluorescent marker or otherwise fluoresce corresponding to their size and position.

Automated Patch Clamp System

Genome editing in the mammalian brain

(Nishiyama, Mikuni, & Yasuda, 2017)

  • Precise genome editing in the brain has historically been challenging because most neurons are postmitotic (non-dividing) and the postmitotic state prevents homology-directed repair (HDR) from occurring. HDR is a mechanism of DNA repair which allows for targeted insertions of DNA fragments with overhangs homologous to the region of interest (by contrast, non-homologous end-joining is highly unpredictable).
  • Nishiyama, Mikuni, and Yasuda developed a technique which allows genome editing in postmitotic mammalian neurons using adeno-associated viruses (AAVs) and CRISPR-Cas9.
  • The AAVs delivered ssDNA sequences encoding a single guide RNA (sgRNA) and an insert. Inserts encoding a hemagglutinin tag (HA) and inserts encoding EGFP were both tested. Cas9 was encoded endogenously by transgenic host cells and in transgenic host animals.
  • The technique achieved precise genome editing in vitro and in vivo with a low rate of off-target effects. Inserts did not cause deletion of nearby endogenous sequences for 98.1% of infected neurons.

Genome Editing Neurons

Near-infrared light and upconversion nanoparticles for optogenetic stimulation

(S. Chen et al., 2018)

  • Upconversion nanoparticles absorb two or more low-energy photons and emit a higher energy photon. For instance, multiple near-infrared photons can be converted into a single visible spectrum photon.
  • Shuo Chen and colleagues injected upconversion nanoparticles into the brains of mice and used them to convert externally applied near-infrared (NIR) light into visible light within the brain tissue. In this way, optogenetic stimulation was performed without the need for surgical implantation of fiber optics or similarly invasive procedures.
  • The authors demonstrated stimulation via upconversion of NIR to blue light (to activate ChR2) and inhibition via upconversion of NIR to green light (to activate a rhodopsin called Arch).
  • As a proof-of-concept, this technology was used to alter the behavior of the mice by activating hippocampally-encoded fear memories.

Upconversion nanoparticles and NIR

Map of all neuronal cell bodies within mouse brain

(Murakami et al., 2018)

  • Ueda, Murakami, and colleagues combined methods from expansion microscopy and CLARITY to develop a protocol called CUBIC-X which both expands and clears entire brains. Light-sheet fluorescence microscopy was used to image the treated brains and a novel algorithm was developed to detect individual nuclei.
  • Although expansion microscopy causes some increased tissue transparency on its own, CUBIC-X greatly improved this property in the enlarged tissues, facilitating more detailed whole-brain imaging.
  • Using CUBIC-X, the spatial locations of all the cell bodies (but not dendrites, axons, or synapses) within the mouse brain were mapped. This process was performed upon several adult mouse brains as well as several developing mouse brains to allow for comparative analysis.
  • The authors made the spatial atlas publicly available in order to facilitate global cooperation towards annotating connectivity among the neural cell bodies within the atlas.

CUBIC-X

Clinical testing of hippocampal prosthesis algorithm in humans

(Hampson et al., 2018)

  • Further clinical tests of Berger’s hippocampal prosthesis were performed. Twenty-one patients took part in the experiments. Seventeen patients underwent CA3 recording so as to facilitate training and optimization of the MIMO model. Eight patients received CA1 stimulation so as to improve their memories.
  • Electrodes with the ability to record from single neurons (10-24 single-neuron recording sites) and via EEG (4-6 EEG recording sites) were implanted such that recording and stimulation could occur at CA3 and CA1 respectively.
  • Patients performed behavioral memory tasks. Both short-term and long-term memory showed an average improvement of 35% across the patients who underwent stimulation.

Precise optogenetic manipulation of fifty neurons

(Mardinly et al., 2018)

  • Mardinly and colleagues engineered a novel excitatory optogenetic ion channel called ST-ChroME and a novel inhibitory optogenetic ion channel called IRES-ST-eGtACR1. The channels were localized to the somas of host neurons and generated stronger photocurrents over shorter timescales than previously existing opsins, allowing for powerful and precise optogenetic stimulation and inhibition.
  • 3D-SHOT is an optical technique in which light is tuned by a device called a spatial light modulator along with several other optical components. Using 3D-SHOT, light was precisely projected upon targeted neurons within a volume of 550×550×100 μm3.
  • By combining novel optogenetic ion channels and the 3D-SHOT technique, complex patterns of neural activity were created in vivo with high spatial and temporal precision.
  • Simultaneously, calcium imaging allowed measurement of the induced neural activity. More custom optoelectronic components helped avoid optical crosstalk of the fluorescent calcium markers with the photostimulating laser.

Optogenetic control of fifty neurons

Whole-brain Drosophila connectome data acquired via serial electron microscopy

(Zheng et al., 2018)

  • Zheng, Bock, and colleagues collected serial electron microscopy data on the entire adult Drosophila connectome, providing the data necessary to reconstruct a complete structural map of the fly’s brain at the resolution of individual synapses, dendritic spines, and axonal processes.
  • The data are in the form of 7050 transmission electron microscopy images (187500 x 87500 pixels and 16 GB per image), each representing a 40nm-thin slice of the fly’s brain. In total the dataset requires 106 TB of storage.
  • Although much of the the data still must be processed to reconstruct a 3-dimensional map of the Drosophila brain, the authors did create 3-dimensional reconstructions of selected areas in the olfactory pathway of the fly. In doing so, they discovered a new cell type as well as several other previously unrealized insights about the organization of Drosophila’s olfactory biology.

Drosophila connectome with SEM

 

References 

Akopyan, F., Sawada, J., Cassidy, A., Alvarez-Icaza, R., Arthur, J., Merolla, P., … Modha, D. S. (2015). TrueNorth: Design and Tool Flow of a 65 mW 1 Million Neuron Programmable Neurosynaptic Chip. IEEE Transactions on Computer-Aided Design of Integrated Circuits and Systems, 34(10), 1537–1557. http://doi.org/10.1109/TCAD.2015.2474396

Berger, T. W., Song, D., Chan, R. H. M., Marmarelis, V. Z., LaCoss, J., Wills, J., … Granacki, J. J. (2012). A Hippocampal Cognitive Prosthesis: Multi-Input, Multi-Output Nonlinear Modeling and VLSI Implementation. IEEE Transactions on Neural Systems and Rehabilitation Engineering, 20(2), 198–211. http://doi.org/10.1109/TNSRE.2012.2189133

Berning, S., Willig, K. I., Steffens, H., Dibaj, P., & Hell, S. W. (2012). Nanoscopy in a Living Mouse Brain. Science, 335(6068), 551 LP-551. Retrieved from http://science.sciencemag.org/content/335/6068/551.abstract

Boyden, E. S., Zhang, F., Bamberg, E., Nagel, G., & Deisseroth, K. (2005). Millisecond-timescale, genetically targeted optical control of neural activity. Nature Neuroscience, 8, 1263. Retrieved from http://dx.doi.org/10.1038/nn1525

Chen, F., Tillberg, P. W., & Boyden, E. S. (2015). Expansion microscopy. Science, 347(6221), 543 LP-548. Retrieved from http://science.sciencemag.org/content/347/6221/543.abstract

Chen, F., Wassie, A. T., Cote, A. J., Sinha, A., Alon, S., Asano, S., … Boyden, E. S. (2016). Nanoscale imaging of RNA with expansion microscopy. Nature Methods, 13, 679. Retrieved from http://dx.doi.org/10.1038/nmeth.3899

Chen, S., Weitemier, A. Z., Zeng, X., He, L., Wang, X., Tao, Y., … McHugh, T. J. (2018). Near-infrared deep brain stimulation via upconversion nanoparticle–mediated optogenetics. Science, 359(6376), 679 LP-684. Retrieved from http://science.sciencemag.org/content/359/6376/679.abstract

Chung, K., & Deisseroth, K. (2013). CLARITY for mapping the nervous system. Nature Methods, 10, 508. Retrieved from http://dx.doi.org/10.1038/nmeth.2481

Constine, J. (2017). Facebook is building brain-computer interfaces for typing and skin-hearing. TechCrunch. Retrieved from https://techcrunch.com/2017/04/19/facebook-brain-interface/

Cyranoski, D. (2017). China launches brain-imaging factory. Nature, 548(7667), 268–269. http://doi.org/10.1038/548268a

Deadwyler, S. A., Hampson, R. E., Song, D., Opris, I., Gerhardt, G. A., Marmarelis, V. Z., & Berger, T. W. (2017). A cognitive prosthesis for memory facilitation by closed-loop functional ensemble stimulation of hippocampal neurons in primate brain. Experimental Neurology, 287, 452–460. http://doi.org/https://doi.org/10.1016/j.expneurol.2016.05.031

Deadwyler, S., Hampson, R., Sweat, A., Song, D., Chan, R., Opris, I., … Berger, T. (2013). Donor/recipient enhancement of memory in rat hippocampus. Frontiers in Systems Neuroscience. Retrieved from https://www.frontiersin.org/article/10.3389/fnsys.2013.00120

Etherington, D. (2017). Elon Musk’s Neuralink wants to boost the brain to keep up with AI. TechCrunch. Retrieved from techcrunch.com/2017/03/27/elon-musks-neuralink-wants-to-boost-the-brain-to-keep-up-with-ai/

Fact Sheet: BRAIN Initiative. (2013). Retrieved from https://obamawhitehouse.archives.gov/the-press-office/2013/04/02/fact-sheet-brain-initiative

Flesher, S. N., Collinger, J. L., Foldes, S. T., Weiss, J. M., Downey, J. E., Tyler-Kabara, E. C., … Gaunt, R. A. (2016). Intracortical microstimulation of human somatosensory cortex. Science Translational Medicine. Retrieved from http://stm.sciencemag.org/content/early/2016/10/12/scitranslmed.aaf8083.abstract

Gunaydin, L. A., Yizhar, O., Berndt, A., Sohal, V. S., Deisseroth, K., & Hegemann, P. (2010). Ultrafast optogenetic control. Nature Neuroscience, 13, 387. Retrieved from http://dx.doi.org/10.1038/nn.2495

Hampson, R. E., Song, D., Robinson, B. S., Fetterhoff, D., Dakos, A. S., Roeder, B. M., … Deadwyler, S. A. (2018). Developing a hippocampal neural prosthetic to facilitate human memory encoding and recall. Journal of Neural Engineering, 15(3), 36014. http://doi.org/10.1088/1741-2552/aaaed7

Han, X., & Boyden, E. S. (2007). Multiple-Color Optical Activation, Silencing, and Desynchronization of Neural Activity, with Single-Spike Temporal Resolution. PLOS ONE, 2(3), e299. Retrieved from https://doi.org/10.1371/journal.pone.0000299

Hatmaker, T. (2017). DARPA awards $65 million to develop the perfect, tiny two-way brain-computer interface. TechCrunch. Retrieved from techcrunch.com/2017/07/10/darpa-nesd-grants-paradromics/

Liu, J., Fu, T.-M., Cheng, Z., Hong, G., Zhou, T., Jin, L., … Lieber, C. M. (2015). Syringe-injectable electronics. Nature Nanotechnology, 10, 629. Retrieved from http://dx.doi.org/10.1038/nnano.2015.115

Livet, J., Weissman, T. A., Kang, H., Draft, R. W., Lu, J., Bennis, R. A., … Lichtman, J. W. (2007). Transgenic strategies for combinatorial expression of fluorescent proteins in the nervous system. Nature, 450, 56. Retrieved from http://dx.doi.org/10.1038/nature06293

Lomas, N. (2017). Superintelligent AI explains Softbank’s push to raise a $100BN Vision Fund. TechCrunch. Retrieved from https://techcrunch.com/2017/02/27/superintelligent-ai-explains-softbanks-push-to-raise-a-100bn-vision-fund/

Mardinly, A. R., Oldenburg, I. A., Pégard, N. C., Sridharan, S., Lyall, E. H., Chesnov, K., … Adesnik, H. (2018). Precise multimodal optical control of neural ensemble activity. Nature Neuroscience, 21(6), 881–893. http://doi.org/10.1038/s41593-018-0139-8

Markram, H. (2006). The Blue Brain Project. Nature Reviews Neuroscience, 7, 153. Retrieved from http://dx.doi.org/10.1038/nrn1848

Markram, H., Muller, E., Ramaswamy, S., Reimann, M. W., Abdellah, M., Sanchez, C. A., … Schürmann, F. (2015). Reconstruction and Simulation of Neocortical Microcircuitry. Cell, 163(2), 456–492. http://doi.org/10.1016/j.cell.2015.09.029

Marx, V. (2013). Neuroscience waves to the crowd. Nature Methods, 10, 1069. Retrieved from http://dx.doi.org/10.1038/nmeth.2695

Murakami, T. C., Mano, T., Saikawa, S., Horiguchi, S. A., Shigeta, D., Baba, K., … Ueda, H. R. (2018). A three-dimensional single-cell-resolution whole-brain atlas using CUBIC-X expansion microscopy and tissue clearing. Nature Neuroscience, 21(4), 625–637. http://doi.org/10.1038/s41593-018-0109-1

Nishiyama, J., Mikuni, T., & Yasuda, R. (2017). Virus-Mediated Genome Editing via Homology-Directed Repair in Mitotic and Postmitotic Cells in Mammalian Brain. Neuron, 96(4), 755–768.e5. http://doi.org/10.1016/j.neuron.2017.10.004

Oizumi, M., Albantakis, L., & Tononi, G. (2014). From the Phenomenology to the Mechanisms of Consciousness: Integrated Information Theory 3.0. PLOS Computational Biology, 10(5), e1003588. Retrieved from https://doi.org/10.1371/journal.pcbi.1003588

Okano, H., Miyawaki, A., & Kasai, K. (2015). Brain/MINDS: brain-mapping project in Japan. Philosophical Transactions of the Royal Society of London. Series B, Biological Sciences, 370(1668). http://doi.org/10.1098/rstb.2014.0310

Poo, M., Du, J., Ip, N. Y., Xiong, Z.-Q., Xu, B., & Tan, T. (2016). China Brain Project: Basic Neuroscience, Brain Diseases, and Brain-Inspired Computing. Neuron, 92(3), 591–596. http://doi.org/10.1016/j.neuron.2016.10.050

Regalado, A. (2017). The Entrepreneur with the $100 Million Plan to Link Brains to Computers. MIT Technology Review. Retrieved from https://www.technologyreview.com/s/603771/the-entrepreneur-with-the-100-million-plan-to-link-brains-to-computers/

Reimann, M. W., Nolte, M., Scolamiero, M., Turner, K., Perin, R., Chindemi, G., … Markram, H. (2017). Cliques of Neurons Bound into Cavities Provide a Missing Link between Structure and Function. Frontiers in Computational Neuroscience. Retrieved from https://www.frontiersin.org/article/10.3389/fncom.2017.00048

Seo, D., Neely, R. M., Shen, K., Singhal, U., Alon, E., Rabaey, J. M., … Maharbiz, M. M. (2016). Wireless Recording in the Peripheral Nervous System with Ultrasonic Neural Dust. Neuron, 91(3), 529–539. http://doi.org/10.1016/j.neuron.2016.06.034

Song, D., She, X., Hampson, R. E., Deadwyler, S. A., & Berger, T. W. (2017). Multi-resolution multi-trial sparse classification model for decoding visual memories from hippocampal spikes in human. In 2017 39th Annual International Conference of the IEEE Engineering in Medicine and Biology Society (EMBC) (pp. 1046–1049). http://doi.org/10.1109/EMBC.2017.8037006

Suk, H.-J., van Welie, I., Kodandaramaiah, S. B., Allen, B., Forest, C. R., & Boyden, E. S. (2017). Closed-Loop Real-Time Imaging Enables Fully Automated Cell-Targeted Patch-Clamp Neural Recording In Vivo. Neuron, 95(5), 1037–1047.e11. http://doi.org/https://doi.org/10.1016/j.neuron.2017.08.011

Szigeti, B., Gleeson, P., Vella, M., Khayrulin, S., Palyanov, A., Hokanson, J., … Larson, S. (2014). OpenWorm: an open-science approach to modeling Caenorhabditis elegans. Frontiers in Computational Neuroscience. Retrieved from https://www.frontiersin.org/article/10.3389/fncom.2014.00137

Zheng, Z., Lauritzen, J. S., Perlman, E., Robinson, C. G., Nichols, M., Milkie, D., … Bock, D. D. (2018). A Complete Electron Microscopy Volume of the Brain of Adult Drosophila melanogaster. Cell, 174(3), 730–743.e22. http://doi.org/10.1016/j.cell.2018.06.019

 

 

 

Some interesting organisms of the ocean


2 Comments

cover image credit: Ryo Minemizu (for more, see https://www.ryo-minemizu.com/)

Anglerfish https://en.wikipedia.org/wiki/Anglerfish

Notes: males are tiny compared to females. In many species of anglerfish, mating occurs through males attaching to and then fusing with females such that their circulatory systems join together. The male provides sperm and is eventually absorbed into the female.

Image: [credit: Dante Fenolio]


Basking shark https://en.wikipedia.org/wiki/Basking_shark

Notes: second largest type of shark (after the whale shark), reaches about 7.9 m in length. They feed on plankton by swimming forwards with the mouth open and filtering via gill rakers.

Image: [credit: Wikipedia page]


Blanket octopus https://en.wikipedia.org/wiki/Blanket_octopus

Notes: characterized by transparent flaps connecting the dorsal and dorsolateral tentacles in adult females. In a case of extreme sexual dimorphism, the females grow to around 2 m in size while the males are only about 2.4 cm.

Image: [credit: Steve Hamedl]


Colossal squid https://en.wikipedia.org/wiki/Colossal_squid

Notes: at around 10 m long, they are shorter than giant squid, but they are much heavier. Colossal squid also has the largest eyes of any organism (around 30-40 cm in diameter).

Image: [credit: NPR]


Crinoid https://en.wikipedia.org/wiki/Crinoid

Notes: depending on the type crinoid adults can either swim freely or be tethered to the sea floor by a stalk. The former are called feather stars and the latter are called sea lillies. Crinoids can also crawl using rootlike structures called cirri as legs. They consume plankton and detritus by filtering through their featherlike arms and then propelling it towards a mouth. They reproduce sexually, releasing sperm and eggs into the water. Fertilized eggs hatch into freely swimming larvae that settle on the sea floor and transition into a stalked juvenile state before eventually breaking away (in the case of feather stars) as adults to swim freely once more.

Image: [credit: Wikipedia page]


Cuttlefish https://en.wikipedia.org/wiki/Cuttlefish

Notes: cuttlefish are among the most intelligent invertebrates. They can rapidly change color using their chromatophore cells as a mode of communication and camouflage as well as to warn off predators (called a deimatic display).

Image: [credit: Wikipedia page]


Garden eels https://en.wikipedia.org/wiki/Heterocongrinae

Notes: garden eels are distinguished by their behavior of living in burrows on the sea floor and poking their heads out of the burrows to eat prey and sliding back into the burrows to avoid predators. Colonies of them can resemble grasses, hence their name.

Image: [credit: Insider]


Glaucus atlanticus https://en.wikipedia.org/wiki/Glaucus_atlanticus

Notes: a type of small (up to 3 cm) sea slug that floats upside down using a gas-filled stomach to stay adrift. They feed on Portuguese man o’ war jellyfish and similar organisms. After consuming venomous nematocysts from their prey, they store the stinging cells in sacs (called cnidosacs) located in their own extremities. This concentrates the nematocysts, making the sting of Glaucus atlanticus potentially more potent even than that of the man o’ war jellyfish itself.

Image: [credit: Wikipedia page]


Hawaiian bobtail squid https://en.wikipedia.org/wiki/Euprymna_scolopes

Notes: enjoys a symbiotic relationship with bioluminescent Aliivibrio fischeri bacteria that inhabit a light organ in the squid’s mantle.

Image: [credit: Wikipedia page]


Japanese spider crab https://en.wikipedia.org/wiki/Japanese_spider_crab

Notes: has the largest legspan of any arthropod, reaching about 3.7 m across.

Image: [credit: Kids Discover]


Lion’s mane jellyfish https://en.wikipedia.org/wiki/Lion’s_mane_jellyfish

Notes: one of the largest types of jellyfish. Though their sizes vary widely, some can reach a bell diameters of more than 2 m and possess tentacles extending for over 30 m in length.

Image: [credit: The Toronto Star]


Placozoa https://en.wikipedia.org/wiki/Placozoa

Notes: a category of animals that exist as flat sheets a few cells thick with a ciliated epithelium on their undersides. They use these cilia to move along the seafloor and most of them reproduce asexually by budding or fragmenting into smaller individuals (though one subtype does also reproduce sexually).

Image: [credit: Wikipedia page]


Prochlorococcus https://en.wikipedia.org/wiki/Prochlorococcus

Notes: a type of marine cyanobacteria which represents perhaps the most abundant photosynthetic organism on Earth.

Image: [credit: Wikipedia page]


Sunflower sea star https://en.wikipedia.org/wiki/Sunflower_sea_star

Notes: large sea stars which can reach diameters of 1 m. Sunflower sea stars are predatory and consume various prey such as sea urchins, other sea stars, clams, sea cucumbers, and more. They move at a speed of about a meter per minute using thousands of tube feet located on their undersides.

Image: [credit: Wikipedia page]


Vampire squid https://en.wikipedia.org/wiki/Vampire_squid

Notes: a deep sea cephalopod of about 30 cm length that lives in the ocean’s aphotic zone. Vampire squid (Vampyroteuthis infernalis) have flaps of tissue connecting their tentacles, each of which is lined by fleshy spines. It is covered in photophores that produce disorientating flashes of light to confuse predators. Rather than ink, they can eject a sticky cloud of bioluminescent mucus when highly agitated by predators.

Image: [credit: Wikipedia page]

Cyborg Earth and the Technological Embryogenesis of the Biosphere


1 Comment

Humongous Fungus, a specimen of Armillaria ostoyae, has claimed the title of world’s largest single organism. Though it features honey mushrooms above ground, the bulk of this creature’s mass arises from its vast subterranean mycelial network of filamentous tendrils. It has spread across more than 2,000 acres of soil and weighs over 30,000 metric tons. Yet I would contend that Humongous Fungus represents a mere microcosm of the world’s true largest organism, a creature that I will call Cyborg Earth. What is Cyborg Earth? Eastern religions have suggested that all life is fundamentally interconnected. Cyborg Earth represents an extension of this concept.

All across the globe, biological life thrives. Quintillions upon quintillions of biomolecular computations happen every second, powering all life. Mycoplasma bacteria. Communities of leafcutter ants. The Humongous Fungus. Beloved beagles. Seasonal influenza viruses. Parasitic roundworms. Families of Canadian elk. Vast blooms of cyanobacteria. Humanity. Life works because of complexity that arises from simplicity that in turn arises from whatever inscrutable quantum mechanical rules lay beneath the molecular scale.

All creatures rearrange atoms in various ways. Termites and beavers rearrange larger bunches of atoms than most organisms. As humans progressed from paleolithic to metalwork to industrialization and then to the space age, information revolution, and era of artificial intelligence, they learned to converse with the atoms around them in an ever more complex fashion. We are actors in an operatic performance, we are subroutines of evolution, we are interwoven matryoshka patterns, an epic chemistry.

Thermodynamics and memetic natural selection juggle our civilization while riding a unicycle along a path to an as-yet unknown destiny. Because of humanity, technology represents a fundamental part of the biosphere. Computers, airplanes, factories, shipping routes. All of it is natural. This does not mean it is good or bad. Simply that it comprises part of the great biological conversation. This is Cyborg Earth. The world’s largest organism is the biosphere itself, including all of the remarkable ways that its constituents have reshaped its body.

Cyborg Earth has always been a uniquely gleaming gem in the cold vastness of the cosmos. But I believe that Cyborg Earth still resides in an embryonic state. As we hurtle into the expanse of the infinite future, Cyborg Earth will grow and change. We must realize that we are all connected by the electric dance of atoms. As a constituent species within Cyborg Earth, we currently possess an enormous power and a tremendous responsibility to properly steward our world’s embryogenesis.

Cover image modified from “Confocal 3D-image of a fungal network with reproductive spores containing nuclei” by Vasilis Kokkoris, source.

Feasibility of mapping the human brain with expansion x-ray microscopy


No Comments

By Logan Thrasher Collins

Click here for PDF version

Combining synchrotron x-ray microscopy with expansion microscopy may represent a feasible approach for whole human brain connectomics at the nanoscale. Synchrotron x-ray microtomography on its own provides extremely fast imaging at high resolution, yet necessary tradeoffs between imaging throughput and resolution mean that the imaging an entire human brain with voxel sizes of less than 100 nm may still take much too long with current synchrotron technology. Fast imaging with voxel sizes of 300-1000 nm is much more readily achievable in the near future. Furthermore, expansion microscopy isotropically enlarges tissue by infusion of a swellable hydrogel, facilitating resolution increases. The combination of x-ray microtomography and expansion microscopy (hereafter referred to as ExxRM) could thus push the effective voxel size down to the level needed for dense connectomics. However, because tissue volume and imaging time scale cubically with expansion factor, careful balance between design of the synchrotron x-ray optical setup and the degree of expansion will be vital. In this perspective, I will explore balances between synchrotron optical engineering choices and expansion factor, propose methods to successfully implement ExxRM in the context of human brains, and estimate how much it would cost to image the human brain in this way. Imaging brains via ExxRM may represent a crucial paradigm shift in connectomics which paves the way for holistic understanding of human brain function.

Introduction

Nanoscale connectomic imaging of the entire human brain represents a long sought-after goal that could provide the foundation for dramatic advances in neurobiology, neurotechnology, and artificial intelligence.1,2 Currently, the leading method for nanoscale connectomics is volume electron microscopy (EM). But imaging a 1 mm3 volume of mouse cortex over a period of 6 months required a tremendous collaborative effort by Yin et al. to develop a parallelized and fully automated transmission electron microscopy (TEM) system consisting of six instruments working in parallel.3 Each of these six instruments cost $125,000. The mouse brain has a volume of roughly 500 mm3, meaning that if these numbers were directly scaled, the process would take 250 years.4 That said, Yin et al.’s TEM dataset had high resolution with 4×4×40 nm voxels, so throughput might be increased by imaging at somewhat lower resolution. As such, it is conceivable to argue that advances in EM technology may enable imaging of an entire mouse brain at sub-100 nm voxel size over the course of a few years at a cost of around $10M. EM therefore represents a viable option for mouse brain connectomics. But the human brain’s volume is roughly 1200 cm3, around 2400-fold larger than the mouse brain.5 Even if EM technology somehow advances to the point where an entire mouse brain can be imaged at 100 nm3 voxel size in a single year for $10M, mapping a human brain with comparable parameters would take thousands of years. In my view, this provides a strong argument for the idea that a radically different approach is necessary for human brain connectomics.

Expansion light-sheet fluorescence microscopy represents a promising alternative to EM, yet this modality also falls short when considering the volume of the human brain (particularly after expansion). For instance, Lillvis et al. utilized 8-fold expansion and lattice light-sheet microscopy (ExLLSM) to image the Drosophila central complex in three colors with effective 30×30×100 nm voxels over the course of 5 days.6 But even accounting for the 8-fold expansion (512-fold volume increase), this amounts to a volume of less than 0.5 mm3. Imaging even a 4-fold expanded mouse brain assuming these numbers would take 876 years. Lattice light-sheet microscopes are relatively inexpensive at a few hundred thousand dollars each7 and thus might be parallelized enough to image an entire mouse brain within a year. However, the 2400-fold larger volume of the human brain relative to the mouse brain indicates that it is probably not reasonable to expect success in human connectomics through ExLLSM.

Based on these calculations, I suggest that a radically different strategy is needed for to put the goal of human brain connectomics within reach. Uniting synchrotron x-ray microtomography (XRM) (Figure 1A-B), expansion microscopy (ExM), a recent staining method known as Unclearing Microscopy8 may facilitate “ExxRM” imaging of the human brain at sub-100 nm voxel size on timescales of around 1 year for a cost of around $10M. Success of this approach will necessitate overcoming some technical hurdles, yet I am optimistic that these particular challenges can be conquered. ExxRM may represent a feasible platform to acquire images suitable for dense connectomics across entire human brains.

Figure 1 Principles of synchrotron XRM. (A) Synchrotrons generate electrons from a source, propel them through a linear accelerator (linac), raise their energy in a booster ring, and then keep the electrons circulating for long periods of time at relativistic speeds in the storage ring. As relativistic electrons move along a curved path controlled by bending magnets, they emit brilliant x-ray beams in the direction tangent to the curve in the direction of the electron movement. Insertion devices that inject the x-rays into beamlines are placed at straight sections of the synchrotron ring. These insertion devices stimulate emission of bright and coherent beams into the experimental stations. (B) Microtomography beamlines receive an x-ray beam from an insertion device and filter out a narrow band of wavelengths using a monochromator. The beam passes through a sample on a rotating tomography stage that can be positionally adjusted to change the location of the field of view within the sample. Projection images are taken across 360° of rotation. X-rays passing through the sample are converted to visible light using a scintillator, then directed by mirrors to a lens system that magnifies the image. This light hits a detector camera and data is recorded for 3D reconstruction.

Recommended methodologies for ExxRM

ExxRM’s success will require developing methodological strategies to mitigate technical challenges. Obtaining sufficient contrast to resolve subcellular features will be vital. ExM cubically dilutes the amount of cellular material per unit volume, so creative staining techniques will be needed. Approaches that ensure stability of expanded tissues under brilliant x-ray illumination for long durations will also be crucial. Multicolor imaging would greatly benefit the usefulness of whole brain connectomics data, so ways of efficiently obtaining tomograms in multiple colors are needed. These challenges must be conquered to translate ExxRM.

A central engineering hurdle for ExxRM is attaining sufficient feature contrast for capture of clear images. Because expanded tissues experience cubic dilution of target biomolecules, contrast from traditional stains such as osmium tetroxide almost entirely vanishes during x-ray microtomographic imaging (Collins et al., unpublished data). An elegant solution to this problem has come in the form of a recently developed technique called Unclearing Microscopy.8 For this technique, M’Saad et al. biotinylated primary amines (found on proteins, phosphatidylethanolamine lipids, etc.) throughout expanded samples, treated the sample with streptavidin horseradish peroxidase (streptavidin-HRP) fusion protein, and then stained with ionic silver reagents (from the EnzMet™ HRP Detection Kit) or with 3’3-diaminobenzidine (DAB). This triggered enzymatic deposition of enough chromogenic silver or DAB to make 20-fold expanded HeLa cells visible to the naked eye despite their 8000-fold increase in volume relative to the unexpanded state. Phase contrast light microscopy subsequently revealed subcellular features such as mitochondrial cristae, nuclear pore complexes, and nuclear membrane. Unclearing thus facilitates physical reconstruction of the structures that are pulled apart by ExM, filling in the gaps left by the expansion process (Figure 2A-B). Silver stain Unclearing could enable either absorption XRM or phase contrast XRM of expanded tissues since silver has excellent x-ray attenuation index β at relevant beam energies as well as excellent x-ray phase decrement δ. Focusing on phase contrast XRM may represent a better option since it can decrease the necessary dose of radiation per unit time by using x-ray wavelengths which are not absorbed as strongly by the tissue. Furthermore, phase contrast XRM is most sensitive to differences in sample density9 and silver staining forms dense precipitates of metallic silver (which in pure form has a high density of 10.49 g/cm3), so this approach might provide superior contrast in the context of ExxRM. Here, ExM’s sample dilution might prove advantageous because it could generate strongly distinct densities between silver-stained cellular features and the rest of the stabilized hydrogel. It should be noted that, for phase contrast XRM, the previously mentioned stabilization approach would need to fill the space between cellular features with a substance that differs substantially in density from the silver (or similar stain). Combining Unclearing Microscopy with phase contrast XRM could provide excellent feature contrast for ExxRM connectomics.

Figure 2 Proposed sample preparation technique for ExxRM. (A) Macroscale view of a human brain undergoing expansion, Unclearing, and stabilization. Slicing into centimeter-scale subvolumes is not shown but may also represent a beneficial strategy. (B) Nanoscale view of a neuron within the brain undergoing expansion, Unclearing, and stabilization. Considerable amplification of signal density after Unclearing should occur.

Expanded tissues are known for their fragility and synchrotron x-rays are known for their harshness, so strategies for solving the problem of sample degradation are needed. Newer expansion recipes which use high monomer concentrations have displayed substantially greater physical sturdiness than earlier generations of ExM hydrogels,10,11 so this may aid in stabilization to some degree. Yet additional advances in ExM sample preparation may still be necessary due to high required fluxes and long imaging times. Cutting human brain samples into smaller volumes on the order of a few centimeters may improve the situation by decreasing imaging times per sample. Keeping the sample at cryogenic temperatures with the help of cryoprotectants to prevent ice crystals damaging the tissue may also help since heat generation from the x-rays is particularly problematic for hydrated samples.12,13 That said, low temperatures would not fix the issue of radiation damage from ionization, so this issue should still be considered.14 Further stabilization might come in the form of infusion of a rapidly crosslinkable or crystalizing substance which irreversibly locks all biomolecules into place (Figure 2A-B). The chosen substance would need to undergo an inducible crosslinking or crystallization reaction that does not cause distortion of the expanded tissue, to not shrink or distort the expanded tissue while it diffuses into the hydrogel, and to minimally absorb x-rays at the energy range used for imaging, and to contrast sharply with the chosen stain material. The phase decrement of the stabilizing substance should be similar or lower compared to amorphous ice. These methods may enable sufficient stabilization for preventing nanoscale tissue degradation even with exposure to extremely brilliant synchrotron x-rays.

Multicolor ExxRM would greatly enhance the value of acquired image data since synapses and key biomolecules could then be tagged within the 3D reconstruction.15,16 Fortunately, multicolor absorption x-ray microscopy represents an established technique wherein two or more staining materials undergo two or more rounds of imaging at beam energies corresponding to the absorption edges of the chosen materials. The absorption edges in question must be sufficiently distinct that minimal overlap in detection occurs across the different beam energy imaging rounds. As an example, Depannemaecker et al. employed this approach by using antibody-linked gold nanoparticles to mark neuronal nuclei in mouse brains while also staining neurons with silver via Golgi’s method.15 They imaged at a beam energy corresponding to an absorption edge of silver and at a beam energy corresponding to an absorption of gold. In addition, multicolor phase contrast x-ray imaging might be accomplished by using molecularly targeted tags with distinct densities relative to the Unclearing silver stain. For instance, gold has a density about twice that of silver, so it might be possible to segment gold nanoparticles after data acquisition. A potential challenge for these methods comes from the difficulty of inducing reliable diffusion of large metallic nanoparticles into tissue. That said, post-expansion pre-stabilization staining could help overcome the problem since expanded gels are typically more porous than pre-expansion tissue. Multicolor ExxRM in 2-3 colors should be achievable, opening doors to more useful whole-brain image datasets.

Though some trial and error will doubtless prove necessary for ExxRM optimization, the required technologies lay within fairly close reach. High contrast could be achieved by applying Unclearing Microscopy to counter signal dilution from expansion. Stability under brilliant x-rays may be possible by leveraging stabler expansion recipes with higher monomer concentrations, by cutting the brain into centimeter-scale subvolumes to decrease radiation dose per sample, by employing cryogenic temperatures during imaging, and by developing rigidity-enhancing materials to infuse into expanded gels. Multicolor ExxRM could be achieved by staining with metallic nanoparticles linked to affinity reagents and performing additional scans tuned to the absorption edges of the chosen metals. These directions may provide a path towards successful ExxRM.

How fast can synchrotrons image expanded brains?

Synchrotron facilities offer bright and coherent x-rays that can rapidly image large volumes of tissue at high resolutions (Figure 1A-B). For example, Bosch et al. employed synchrotron XRM at the Swiss Light Source (SLS) and the Diamond Light Source (DLS) to acquire 3D reconstructions of an entire mouse olfactory bulb with 325 nm voxel size.17 In their study, tomograms of approximately 1 mm3 volume took around 20 minutes to acquire. Another investigation by Dyer et al. used the Advanced Photon Source (APS) synchrotron to image a region of mouse cortex at 650 nm voxel size and 1.47 mm3 volume. Imaging this volume took only 6 minutes.18 Walsh et al. leveraged the European Synchrotron Radiation Facility Extremely Brilliant Source (ESRF-EBS) to image a variety of whole human organs at low resolution and subvolumes within human organs at higher resolution.19 Notably, they reconstructed a cylinder with ~57 mm3 volume (5.4 mm diameter and 2.5 mm height) within human spleen at 1290 nm voxel size over the course of 2 hours. Ding et al. used the APS to image larval zebrafish with 743 nm voxels and 1.5 mm3 volumes per tomogram in 20 minutes with monochromatic x-ray scans and in just 20 seconds with polychromatic “pink beam” x-ray scans.20 Despite their much higher flux and correspondingly much shorter imaging times, the polychromatic scans unfortunately showed lower resolution due to poor SNR. It should be noted that resolution represents a distinct concept from voxel size and that contrast and SNR play major roles in the final resolution. That said, voxel size can act as a rough proxy for comparisons across different imaging setups assuming that contrast and SNR are consistent. In another key example, Foxley et al. utilized the APS to image whole mouse brains (11.7×11.7×17.7 mm) over the course of 7 hours of acquisition time with 1117 nm voxels.21 Finally, Rodgers et al. used the SOLEIL synchrotron and an alternative technique for tomographic acquisition (discussed in more detail later) to image an entire mouse brain a 650 nm voxel size in 8 hours.22 Though the parameters for image acquisition vary considerably across different biological samples and synchrotron hardware setups (table 1), these examples illustrate the power of synchrotron imaging for mapping biological tissues.

Table 1 Specifications of synchrotron tissue imaging experiments from selected references. 

ReferenceSmallest voxel sizeSingle tomogram volumeTotal reconstructed volumeImaging time per tomogramTotal imaging time
Bosch et al.325 nm1.39 mm3~5-10 mm320 min.~40 min. to ~4 hours
Dyer et al.650 nm1.47 mm31.47 mm36 min.6 min.
Walsh et al.1290 nm57 mm357 mm3120 min.2 hours
Ding et al.743 nm1.5 mm34.5 mm320 min. or 20 s*1 hour or* 1 min.
Foxley et al.1117 nm5.61 mm32423 mm3108 s7 hours
Rodgers et al.650 nmspecial method911 mm3special method8 hours
*with pink beam acquisition

While the voxel sizes described above are not small enough for nanoscale connectomics, incorporating ExM into the sample preparation pipeline presents the possibility of attaining sufficient resolutions for tracing neurites. Expansion factors of around 4x, 10x, and 20x are respectively possible with standard ExM,23,24 some of the newer enhanced ExM protocols,10,25,26 and iterative ExM methods.11,27 Expansion factor can also be intentionally decreased somewhat by adjusting salt concentration. It is important to note that not all ExM protocols retain lipid membranes, so selecting a recipe that preserves such cellular boundaries10,11,28 will be vital for morphology reconstrction. Balance between expansion factor and imaging time will represent a key determinant of the success of ExxRM for whole brain imaging. Higher expansion factors increase resolution linearly while increasing imaging time cubically. Synchrotron imaging’s extreme speed has the potential to keep up with such volumetric increases, though even this has certain limits.

Du et al. offer a helpful mathematical model to estimate the number of photons needed for imaging a sample with sufficient contrast to distinguish between voxels of desired size.12 The model starts by considering how many photons n̄pixel are needed to image a single pixel on a 2D tomographic slice. The pixel is assumed to contain a “feature” material with a phase decrement δf that must be distinguished against a background with a phase decrement δb. Distinctions can be made between adjacent background pixels b, overlaying background pixels bo’ and underlaying background pixels bu’. The SNR is assumed to equal 5 as based on the Rose criterion, which qualitatively represents an acceptable SNR for visually useful images. Background attenuation coefficient is given by µb’ and the thicknesses of the overlaying and underlaying background material are tb’o and tb’u respectively. After the value of n̄pixel is determined, the value is multiplied by the square N2 of the number of pixels N on the detector’s horizontal axis to find the number of photons needed per tomogram. As Du et al. explain, the number of slices is not used as a multiplicative factor since the photons are distributed across all of the rotation angles for each pixel. These calculations act as a guide to what sample properties and beam properties are needed for ExxRM.

The Du et al. model can help determine the feasibility of ExxRM by estimating the necessary synchrotron flux for imaging the brain at a desired voxel size in a reasonable time frame. Consider 1 cm3 of brain tissue expanded 4-fold to a block with dimensions of 4×4×4 cm. Based on this, assume that the average thickness of overlaying and underlaying background material is 2 cm. Also assume that the background material is made of amorphous water ice with a density of 0.92 g/cm3. Since the widths of the smallest neuronal features are about 20 nm wide,29 assume that the 4-fold expansion provides a minimum feature thickness of 80 nm. By using the Unclearing Microscopy technique, such features might be “filled in” after expansion with a metallic silver stain.8 Thus, I will assume that the 4-fold expanded membrane features are made up of metallic silver with a density of 5.25 g/cm3, half the density of pure metallic silver (to account for imperfect staining). Feature phase decrements δf, background phase decrements δb, and background attenuation coefficients µb’ across x-ray energies ranging from 10 keV to 30 keV can be obtained using the calculator available at (https://henke.lbl.gov/optical_constants/pert_form.html) which is based on the Henke dataset.30 This calculator does not directly give µb’, but it does give attenuation index β, for which µb’ = 4πβ/λ. I will furthermore assume that a 3×3 mm beam and detector are possible to employ and that the pixel size is 300 nm (75 nm effective post-expansion). According to the resulting model, the total number of photons needed to image a single tomogram (volume of 21.21 mm3) within the 4-fold expanded block at an energy of 30 keV is 1.47×1013. By examining this model across a range of x-ray energies (Figure 3A) and across a range of feature sizes (Figure 3A), one can obtain a better understanding of how the different variables might play out in an experimental setting.

Figure 3 (A) Total photons needed to image a single tomogram of 4-fold expanded tissue with volume of 21.21 mm3 at 300 nm voxel size and two different silver stain densities while assuming feature thickness of 80 nm, plotted against x-ray energy in keV. (B) Total photons needed to image a single tomogram of 4-fold expanded tissue with volume of 21.21 mm3 at 300 nm voxel size and two different silver stain densities while assuming 30 keV x-ray energy, plotted against desired feature thickness necessary to resolve.

How might this translate to imaging time for a single tomogram, the 4×4×4 cm expanded block, and the whole human brain? Contemporary beamlines which employ multilayer monochromators can reach fluxes of more than 1012 photons/s.31–35 As such, the single tomogram under the assumptions of the model might be acquired in around 10 seconds. Comparing the tomogram volume of 21.21 mm3 and the expanded block volume of 64000 mm3, this means that the whole block would require about 3000 tomograms and take 8.3 hours to image. Since the entire human brain should contain approximately 1200 of these blocks, whole-brain acquisition might take around 1.14 years of continuous imaging. All of this assumes the post-expansion effective voxel size of 75 nm (4-fold expansion and actual voxel size of 300 nm), 30 keV beam energy, and Unclearing of neuronal features to a density of 5.25 g/cm3 metallic silver. If these theoretical projections successfully align with experimental outcomes, ExxRM could facilitate nanoscale connectomics for the entire human brain in a time frame of around 1-2 years.

Cost estimates for whole brain ExxRM

While the power of the synchrotron facility comes with a high price tag, ExxRM may still represent the most overall cost-effective option for human brain connectomics. Consider the costs associated with the DLS as an illustrative example. The DLS is a third-generation facility and is currently one of the better synchrotrons in terms of its ability to produce bright and coherent x-rays. Building the DLS and its first seven beamlines from 2003-2007 cost about $316M, its later upgrades cost $144M and $134M, and its yearly operational costs have increased from $28M in 2007-2008 to $81M in 2019-2020 (table 2). Based on these data points, construction of a new synchrotron beamline costs approximately $10M and yearly maintenance may cost roughly $500K. This provides a framework for estimating the cost of a dedicated human brain ExxRM connectomics beamline.

Table 2 Costs associated with the Diamond Light Source36 as a case study on how much money is needed to build and maintain a state-of-the-art synchrotron facility.

FacilityDiamond Light Source
Initial costs$316M: synchrotron, first seven beamlines, surrounding buildings, construction started in 2003 and completed in 2007
Upgrade 1 costs$144M: fifteen more beamlines, detector development program, construction started in 2004 and completed in 2012
Upgrade 2 costs$134M: ten more beamlines, not stated when construction started but was completed in 2021
Maintenance and operational costs$28M in 2007-2008, $48M in 2012-2013, $81M in 2019-2020

Compact Light Source (CLS) technology should be considered as well before continuing. CLS instruments produce x-ray beams that fall somewhere between laboratory x-ray microscopes and synchrotrons in terms of brightness and coherence.37 CLS instruments are furthermore small enough to fit into a single room and are inexpensive enough that a large number of them could potentially be constructed in parallel. At first glance, CLS technology seems a more economically viable alternative to synchrotron beamlines, yet it still probably is not a good option at this time. Existing CLS instruments are not likely suitable for human brain (or even mouse brain) ExxRM connectomics in the foreseeable future because their optical engineering requirements and mediocre level of x-ray flux preclude rapid tomography at submicron resolution, particularly when a large field of view is desired.38 There is a small possibility that future advances in CLS technology could change this situation, yet this seems fairly unlikely, so synchrotron-based imaging remains the best route.

Building an entire synchrotron solely for ExxRM connectomics is probably less efficient than establishing an agreement with an existing synchrotron to construct a connectomics beamline. While one might envision additional parallelization through custom design of the beamline to split the beam to land in multiple sample chambers, splitting the beam would divide the photon flux and therefore increase imaging times for no net gain in speed. As such, parallelization would likely require an additional insertion device and thus an entire new beamline for each new sample chamber. Insertion devices consist of a series of precisely engineered magnets built into straight sections of the synchrotron’s ring. These magnets, known as undulators or wigglers depending on the specifics of the insertion device, stimulate directed emission of a brilliant x-ray beam out from the storage ring.39 Yet even if we assume $10M total plus yearly maintenance costs for each connectomics beamline, imaging multiple human brains over the course of a year or a single human brain in just a few months remains a reasonable proposition. For a project as important as mapping the human brain at sufficient resolution for dense neuronal reconstruction, price tags in the range of tens of millions of dollars may not be out of reach.

Conclusion and outlook

Connectomics needs a technological paradigm shift if it is to feasibilize dense mapping of one or more human brains. Though it comes with some technical challenges, ExxRM may represent a key strategic shift that drastically reduces human brain connectomics timelines from centuries down to 1-2 years for the image acquisition step. Data storage and early processing steps (e.g. tomographic reconstruction) will of course require data centers and high-performance computing, but this field is rapidly advancing and may indeed be capable of handling the challenge. Assuming 1 byte per voxel, the amount of storage needed for a 4-fold expanded human brain with 300 nm physical voxel size (75 nm effective voxel size) is about 2.84 exabytes. Segmentation of the human brain dataset will probably represent a vastly more difficult problem as well as require substantially more compute resources, so further advances in this area will need to occur in parallel with ExxRM development. Realizing the benefits of connectomics in the form of complete computational models of the brain will take additional extensive research. Precisely correlating gene expression and electrophysiological properties with neuronal morphology (i.e. “cell type”) may represent a major step towards bridging the divide between structural data and functional activity. Nonetheless, the allure of having a holistic anatomical picture of the brain may serve as a driving force in the meantime. ExxRM has the potential to transform the dense connectomics field, enabling anatomical imaging of the entire human brain with sub-100 nm voxel size and high contrast in around 1-2 years for a price of roughly $10M.

References

1.        Collins, L. T. The case for emulating insect brains using anatomical “wiring diagrams” equipped with biophysical models of neuronal activity. Biol. Cybern. 113, 465–474 (2019).

2.        Koene, R. A. Fundamentals of whole brain emulation: state, transition, and update representations. Int. J. Mach. Conscious. 04, 5–21 (2012).

3.        Yin, W. et al. A petascale automated imaging pipeline for mapping neuronal circuits with high-throughput transmission electron microscopy. Nat. Commun. 11, 4949 (2020).

4.        Badea, A., Ali-Sharief, A. A. & Johnson, G. A. Morphometric analysis of the C57BL/6J mouse brain. Neuroimage 37, 683–693 (2007).

5.        Cosgrove, K. P., Mazure, C. M. & Staley, J. K. Evolving Knowledge of Sex Differences in Brain Structure, Function, and Chemistry. Biol. Psychiatry 62, 847–855 (2007).

6.        Lillvis, J. L. et al. Rapid reconstruction of neural circuits using tissue expansion and light sheet microscopy. Elife 11, e81248 (2022).

7.        Watkins, S. C. & St. Croix, C. M. Light sheet imaging comes of age. J. Cell Biol. 217, 1567–1569 (2018).

8.        M’Saad, O., Shribak, M. & Bewersdorf, J. Unclearing Microscopy. bioRxiv 2022.11.29.518361 (2022) doi:10.1101/2022.11.29.518361.

9.        R A Lewis. Medical phase contrast x-ray imaging: current status and future prospects. Phys. Med. Biol. 49, 3573 (2004).

10.      Klimas, A. et al. Magnify is a universal molecular anchoring strategy for expansion microscopy. Nat. Biotechnol. (2023) doi:10.1038/s41587-022-01546-1.

11.      M’Saad, O. & Bewersdorf, J. Light microscopy of proteins in their ultrastructural context. Nat. Commun. 11, 3850 (2020).

12.      Du, M. et al. Upscaling X-ray nanoimaging to macroscopic specimens. J. Appl. Crystallogr. 54, 386–401 (2021).

13.      Matsuyama, S. et al. Elemental mapping of frozen-hydrated cells with cryo-scanning X-ray fluorescence microscopy. X-Ray Spectrom. 39, 260–266 (2010).

14.      Lombi, E. et al. Fast X-Ray Fluorescence Microtomography of Hydrated Biological Samples. PLoS One 6, e20626 (2011).

15.      Depannemaecker, D. et al. Gold Nanoparticles for X-ray Microtomography of Neurons. ACS Chem. Neurosci. 10, 3404–3408 (2019).

16.      Logan Thrasher Collins, Kayla Siletti, Kristine Fischenich, Jennifer Coulombe, Nathan Anderson, M. S. Structural brain mapping using antibody-conjugated gold nanoparticles and x-ray microscopy. in Society for Neuroscience Conference (2019).

17.      Bosch, C. et al. Functional and multiscale 3D structural investigation of brain tissue through correlative in vivo physiology, synchrotron microtomography and volume electron microscopy. Nat. Commun. 13, 2923 (2022).

18.      Dyer, E. L. et al. Quantifying Mesoscale Neuroanatomy Using X-Ray Microtomography. eNeuro 4, ENEURO.0195-17.2017 (2017).

19.      Walsh, C. L. et al. Imaging intact human organs with local resolution of cellular structures using hierarchical phase-contrast tomography. Nat. Methods 18, 1532–1541 (2021).

20.      Ding, Y. et al. Computational 3D histological phenotyping of whole zebrafish by X-ray histotomography. Elife 8, e44898 (2019).

21.      Foxley, S. et al. Multi-modal imaging of a single mouse brain over five orders of magnitude of resolution. Neuroimage 238, 118250 (2021).

22.      Rodgers, G. et al. Mosaic microtomography of a full mouse brain with sub-micron pixel size. in Proc.SPIE vol. 12242 122421L (2022).

23.      Chen, F., Tillberg, P. W. & Boyden, E. S. Expansion microscopy. Science (80-. ). 347, 543 LP – 548 (2015).

24.      Tillberg, P. W. et al. Protein-retention expansion microscopy of cells and tissues labeled using standard fluorescent proteins and antibodies. Nat. Biotechnol. 34, 987–992 (2016).

25.      Damstra, H. G. J. et al. Visualizing cellular and tissue ultrastructure using Ten-fold Robust Expansion Microscopy (TREx). Elife 11, e73775 (2022).

26.      Park, H.-E. et al. Scalable and Isotropic Expansion of Tissues with Simply Tunable Expansion Ratio. Adv. Sci. 6, 1901673 (2019).

27.      Chang, J.-B. et al. Iterative expansion microscopy. Nat. Methods 14, 593–599 (2017).

28.      Karagiannis, E. D. et al. Expansion Microscopy of Lipid Membranes. bioRxiv 829903 (2019) doi:10.1101/829903.

29.      Sneve, M. A. & Piatkevich, K. D. Towards a Comprehensive Optical Connectome at Single Synapse Resolution via Expansion Microscopy   . Frontiers in Synaptic Neuroscience   vol. 13 at https://www.frontiersin.org/articles/10.3389/fnsyn.2021.754814 (2022).

30.      Henke, B. L., Gullikson, E. M. & Davis, J. C. X-Ray Interactions: Photoabsorption, Scattering, Transmission, and Reflection at E = 50-30,000 eV, Z = 1-92. At. Data Nucl. Data Tables 54, 181–342 (1993).

31.      Albrahim, M. et al. Reduction and Agglomeration of Supported Metal Clusters Induced by High-Flux X-ray Absorption Spectroscopy Measurements. J. Phys. Chem. C 125, 11048–11057 (2021).

32.      Owen, R. L., Holton, J. M., Schulze-Briese, C. & Garman, E. F. Determination of X-ray flux using silicon pin diodes. J. Synchrotron Radiat. 16, 143–151 (2009).

33.      Weitkamp, T. et al. Parallel‐beam imaging at the ESRF beamline ID19: current status and plans for the future. AIP Conf. Proc. 1234, 83–86 (2010).

34.      Baussens, O. et al. Characterization of High-Flux CdZnTe with optimized electrodes for 4th generation synchrotrons. J. Instrum. 17, C11008 (2022).

35.      Moosmann, J. et al. X-ray phase-contrast in vivo microtomography probes new aspects of Xenopus gastrulation. Nature 497, 374–377 (2013).

36.      Diamond Light Source About Us. https://www.diamond.ac.uk/Home/About/ (2022).

37.      Gradl, R. et al. Propagation-based Phase-Contrast X-ray Imaging at a Compact Light Source. Sci. Rep. 7, 4908 (2017).

38.      Gunther, B. et al. The versatile X-ray beamline of the Munich Compact Light Source: design, instrumentation and applications. J. Synchrotron Radiat. 27, 1395–1414 (2020).

39.      Willmott, P. Synchrotron Physics. in An Introduction to Synchrotron Radiation 39–86 (2011). doi:https://doi.org/10.1002/9781119970958.ch3.

Challenges of Particular Interest to Me


1 Comment

As a scientist, I am driven by the power of technological breakthroughs to make positive change for humanity. While I also take immense pleasure in the artistic/creative aspects of technology design, my motivation is centered on helping people and on protecting the future of the human species. For this reason, I am interested in a wide array of contemporary challenges as described in this outline. Because I am a synthetic biologist and synthetic biology has many applications, I have the ability to explore solutions to such diverse challenges despite their highly multidisciplinary nature.

That said, one of the tools in any good researcher’s repertoire is collaboration. Since I am just one person, my knowledge can only go so deep in so many areas. Interdisciplinary projects are much more likely to succeed when experts from multiple areas work together. So, I leverage collaboration extensively when carrying out my projects and will continue to do so in the future.

It should be noted that, though I am publicly presenting a number of conceptual explanations of possible solutions to important problems via this list, I have deliberately stated them in somewhat vague language to prevent their public disclosure from precluding outside investment.

I am sharing this list as a way of increasing my likelihood of connecting with potential collaborators over time and as a method of inspiring others to consider how they too might contribute to building a bright future.

I chose the featured image of a Martian-hued rendering of the Anasazi Cliff Palace at Mesa Verde National Park as a symbolic tribute to the creative and culturally rich spirit of humanity, an homage to where we have come from, and a hopeful indication of where we might go in the future.

Infectious Disease Burden in Developing Countries

  • Especially malaria, tuberculosis, and HIV.
  • Translational strategies for dissemination are key.
  • Some possible solutions:
    • Gene drives to prevent mosquitos from carrying pathogens
    • Inexpensive home diagnostics
    • Rapid inexpensive biomanufacturing of treatments
    • Thermostable treatments and vaccines
    • Inexpensive immune enhancement gene therapies
    • Rapid inexpensive biomanufacturing of vaccines

Antibacterial Resistance

  • Especially highly concerning pathogens such as carbapenem-resistant Enterobacterales, carbapenem-resistant Acinetobacter, Clostridioides difficile, and drug-resistant Neisseria gonorrhoeae.
  • Adaptability and rapid generation of new solutions are key.
  • Some possible solutions:
    • Adaptable probiotic treatments
    • Rational design of phage therapies
    • Diversified phage therapy production platforms
    • Lysogenic phage therapies for widespread resistance shutdown and/or bactericide
    • Sentinel bacteria for detection and elimination of resistance in environment (esp. livestock and wastewater)
    • Bacterial conjugation for elimination of resistance in environment
    • Immune enhancement gene therapies
    • Rapid inexpensive biomanufacturing of vaccines
    • Rapid vaccine discovery platforms
    • Inexpensive home diagnostics

Affordable Gene Therapy Manufacturing

  • Many existing gene therapy delivery vehicles are extremely expensive to produce, so treatments are very costly for patients and not enough can be made to reach large populations.
  • Novel manufacturing solutions are vital to scalably make existing vectors (esp. AAVs).
  • New vectors which retain the benefits of existing vectors yet can be made inexpensively are also important.
  • Some possible solutions:
    • Synthetic biology methods for radically redesigning cellular platforms of virus production
    • Novel inexpensively manufacturable viral vectors
    • Hybrid nanoparticle-viral vectors that are easier to produce yet retain benefits of viruses and perhaps bring extra advantages as well
    • Methods for production of DNA origami nanorobots
    • Use of DNA origami to support viral capsid assembly and genome packaging

Gene Therapy for Radiation Resistance in Space

  • Extended periods of time in space, on the moon, on Mars, etc. expose people to large amounts of radiation.
  • Future space colonization efforts could be severely jeopardized by human radiation exposure, especially since this can cause problems with reproduction.
  • Some possible genetic approaches:
    • Add genes derived from radiation-resistant organisms (e.g. tardigrades, Deinococcus radiodurans, etc.)
    • Enhance human DNA repair pathways by adding new genetic circuits and/or altering gene regulation
    • Polygenic gene therapy delivery vectors
    • Develop ways of safely delivering genes to most or all cells in the human body

Gene Therapy for Aging

  • Aging affects everyone and is marked by a deterioration of health and eventual death.
  • Treatments for aging would greatly improve the human condition by making people both much healthier and longer lived.
  • Life extension only linearly affects population growth, whereas reproduction exponentially affects population growth, so concerns about life extension causing overpopulation are often exaggerated.
  • As human longevity increases, its small contribution to population growth will likely be mitigated by parallel growth of technologies that improve human sustainability (e.g. vertical farms, cultured meat, renewable energy, space habitation, etc.)
  • Gene therapy has great potential for extending human longevity and simultaneously improving overall global health.
  • Some possible genetic approaches:
    • Polygenic gene therapy delivery vectors
    • Identify multiple genes that synergistically improve healthspan and deliver them together
    • Engineer regulatory pathways and insert genetic circuits that optimally modulate gene expression for improving healthspan

Biological Carbon Capture for Climate Change

  • The climate crisis threatens human and nonhuman life since is leading to desertification, flooding, extreme weather events, ecosystem collapse, etc. and will cause many millions of deaths if it continues unchecked.
  • Addressing climate change will necessitate both policy solutions and technological solutions.
  • Carbon capture is a particularly promising route towards mitigating climate change yet is difficult to scale.
  • Self-replicating biological carbon capture approaches would come with risks but demonstrate much greater scalability.
  • Some possible biological carbon capture solutions:
    • Genetically enhanced trees that more rapidly capture carbon
    • Genetically enhanced cyanobacteria (or algae) that more rapidly capture carbon
    • Bacteriophages that propagate genes in ocean cyanobacteria to enhance their carbon uptake
    • Use bacterial conjugation in ocean cyanobacteria to propagate genes that enhance their resistance to bacteriophages and thus rapidly increase cyanobacterial population size and carbon capture capacity
    • Develop computational models and experimental model systems to explore possible negative side effects of all of the above, find ways to counterbalance those side effects

Strong Nanorobotics

  • Strong nanorobots with capabilities resembling those in science fiction would revolutionize every human activity.
  • Strong nanorobots should specifically possess (1) inexpensive mass producibility or self-replication capabilities, (2) ways of programming them to alter their functionality and environmental responses, (3) automated locomotion oriented towards accomplishing programmed tasks.
  • Only relatively simple nanorobots have been created so far.
  • Some possible routes towards strong nanorobotics:
    • Develop minimal cells derived from Mycoplasma bacteria, equip them with extensive optogenetic equipment for external programming, keep the “programming mode” turned off except when a special chemical switch is flipped to prevent light from scrambling their instructions
    • Synthesize complex dynamic DNA origami nanostructures that include optically programmable logic systems inspired by digital logic circuits as well as automated locomotion modules, also devise scalable manufacturing methods

Horizontal Gene Drives to Repair Pollinator Insect Networks

  • Bees and other insect pollinators form a crucial part of global ecosystems, yet populations of these insects are declining.
  • Decline of insect pollinators is negatively affecting many crops, limiting food production across the world.
  • Some of the most prominent reasons for bee decline are the spread of invasive Varroa mites that carry deformed wing virus (DWV) and the prevalence of toxic pesticides in the environment.
  • Horizontal gene transfer via seeding donor bacteria into insect gut microbiota may help protect insect pollinators in a scalable fashion.
  • Some possible approaches involving horizontal gene drives:
    • Give bees gut bacteria that spread conjugative plasmids carrying anti-Varroa genes.
    • Give bees gut bacteria that spread conjugative plasmids carrying anti-DWV genes.
    • Give bees gut bacteria that spread conjugative plasmids carrying genes that facilitate breakdown of toxic pesticides.

Connectomics Towards Whole-Brain Emulation

  • Mapping the brain and simulating it in a computer would provide an unparalleled holistic understanding of how our minds work.
  • Even partial connectomes and/or animal connectomes could give remarkable insights into neurobiology.
  • The convergence of connectomics and computational neuroscience has applications in bioinspired artificial intelligence, bioinspired robotics, neural prostheses, medicine for brain disease, and more.
  • Some possible routes for connectomics:
    • Expansion microscopy with genetically encoded synaptic and neuronal barcodes coupled with spatial transcriptomics
    • Massively parallel electron microscopy approaches
    • Improved x-ray nanotomography coupled with special sample treatment methods to improve tissue stability, allow multicolor imaging (and possibly barcodes), and enhance resolution
    • Construction of many compact light source devices to rapidly map tissue in parallel
    • Construction of extremely bright next-generation synchrotrons coupled with sample treatment methods to greatly improve stability

Nanobiotechnology for Neural Interfaces and Neural Prostheses

  • Existing neural interfaces and neural prostheses utilize microelectrode-based technologies for communicating with brain tissue.
  • Nanobiotechnology approaches may enable more precise, less invasive, and more powerful neural interfaces and prostheses.
  • Some possible approaches:
    • Polymersome nanocompartments that mimic neurons in their electrical response properties via embedded transmembrane proteins and ion gradients, link to nanowires that transmit electrical potential to external devices or other parts of the brain
    • Leverage gene therapy for modifying human neurons to express optogenetic channels, design nanomachines that incorporate upconversion nanoparticles for targeted stimulation

Terraforming

  • The future of humankind depends on our ability to colonize other planets, moons, etc.
  • Earth is the only planet in the solar system where humans can survive unaided.
  • Terraforming the moon or Mars would provide humankind with a new habitable world, yet this represents an enormously challenging task.
  • Seeding the moon or Mars with heavily engineered microorganisms may provide a first step towards transforming these celestial bodies into habitable worlds.
  • Some possible microorganism-based terraforming methods:
    • Perform extensive metabolic engineering on extremophiles that metabolize substances similar to those found on the moon or Mars, make them convert regolith to Earthlike atmospheric gases
    • Borrow genetic pathways from Deinococcus radiodurans to provide radiation resistance
    • Extensively engineer microorganisms to metabolize regolith and form seaweed-like colonies that bud into edible fruits

The Virus Zoo: A Primer on Molecular Virology


2 Comments

Click here for a PDF version of the virus zoo

 

Human Immunodeficiency Virus (HIV)

Genome and Structure:

HIV’s genome is a 9.7 kb linear positive-sense ssRNA.1 There is a m7G-cap (specifically the standard eukaryotic m7GpppG as added by the host’s enzymes) at the 5’ end of the genome and a poly-A tail at the 3’ end of the genome.2 The genome also has a 5’-LTR and 3’-LTR (long terminal repeats) that aid its integration into the host genome after reverse transcription, that facilitate HIV genetic regulation, and that play a variety of other important functional roles. In particular, it should be noted that the integrated 5’UTR contains the HIV promoter called U3.3,4

HIV’s genome translates three polyproteins (as well as several accessory proteins). The Gag polyprotein contains the HIV structural proteins. The Gag-Pol polyprotein contains (within its Pol component) the enzymes viral protease, reverse transcriptase, and integrase. The Gag-Pol polyprotein is produced via a –1 ribosomal frameshift at the end of Gag translation. Because of the lower efficiency of this frameshift, Gag-Pol is synthesized 20-fold less frequently than Gag.5 The frameshift’s mechanism depends upon a slippery heptanucleotide sequence UUUUUUA and a downstream RNA secondary structure called the frameshift stimulatory signal (FSS).6 This FSS controls the efficiency of the frameshift process.

The HIV RNA genome undergoes alternative splicing to produce the rest of the viral proteins. One splicing event produces an RNA that separately encodes the Vpu protein and the Env protein (also called gp160).6–8 A mechanism called ribosome shunting is used to transition from Vpu’s open reading frame to Env’s open reading frame. The Env protein contains the gp41 and gp120 proteins. Env is post-translationally cleaved into gp41 and gp120 by a host furin enzyme in the endoplasmic reticulum.9 It is important to note that Env is also heavily glycosylated post-translationally to help HIV evade the immune system. Several other complex splicing events lead to the production of RNAs encoding Tat, Rev, Nef, Vif, and Vpr.

HIV viral protease cleaves the Gag polyprotein and thus produces structural proteins including the capsid protein CA (also called p24), the matrix protein MA (also called p17), the nucleocapsid protein NC (also called p7), and the p6 peptide.10 The HIV core capsid is shaped like a truncated cone and consists of about 1500 CA monomers. Most of the CA proteins assemble into hexamers, but a few pentamers are present. The pentamers help give the core capsid its conical morphology by providing extra curvature near the top and bottom. Each core capsid contains two copies of the HIV genomic RNA, complexed with NC protein. Reverse transcriptase, integrase, and viral accessory proteins are also held within the core capsid. HIV’s core capsid is packaged into a lipid envelope that bears gp41-gp120 glycoprotein heterodimers. The MA protein forms a layer between the core capsid and the envelope.

Accessory proteins Vpu, Tat, Rev, Nef, Vif, and Vpr facilitate a variety of functions. Vpu induces degradation of CD4 proteins within the endoplasmic reticulum of host CD4+ T cells. It does this by using its cytosolic domain as a molecular adaptor between CD4 and a ubiquitin ligase (which subsequently triggers proteosomal degradation of the CD4).11 The reason that Vpu does this is to prevent HIV superinfection wherein two different types of HIV might infect the same cell and interfere with each other. This is an example of competition between viruses.12 Vpu also enhances release of HIV virions from infected cells by using its cytosolic domain to inhibit a host protein called tetherin (also known as BST-2).11 Without Vpu, tetherin would bind the viral envelope to the cell surface as well to other HIV virus particles, impeding release.

Tat, also called the viral transactivator protein, is necessary for efficient transcriptional elongation of the HIV genome after integration into the host DNA.13 Tat binds the viral transactivation response element (TAR), a structured RNA motif present at the beginning of the HIV transcripts. It then recruits protein positive transcription elongation factor b (P-TEFb). This allows P-TEFb to phosphorylate certain residues in the C-terminal domain of RNA polymerase II, stimulating transcriptional elongation. Tat also recruits several of the host cell’s histone acetyltransferases to the viral 5’-LTR so as to open the chromatin around the U3 promoter and related parts of the integrated HIV genome.3,4 Finally, Tat is secreted from infected cells14 and acts as an autocrine and paracrine signaling molecule.4 It inhibits antigen-specific lymphocyte proliferation, stimulates expression of certain cytokines and cytokine receptors, modulates the activities of various host cell types, causes neurotoxicity in the brain, and more.

Rev facilitates nuclear export of the unspliced and singly spliced HIV RNAs by binding to a sequence located in the Env coding region called the Rev response element (RRE).13 The Rev protein forms a dimer upon binding to the RRE and acts as an adaptor, binding a host nuclear export factor called CRM1. Rev is also known to form higher-order oligomers via cooperative multimerization of the RNA-bound dimers.

Nef is a myristoylated protein that downregulates certain host T cell proteins and thereby increases production of virus. Nef is localized to the cytosol and the plasma membrane. It specifically inhibits CD4, Lck, CTLA-4, and Bad.15 Downregulating CD4 contributes to the prevention of superinfection that also occurs with Vpu’s inhibition of CD4. Nef induces endocytosis of plasma membrane Lck protein and traffics it to recycling endosomes and the trans-Golgi network. At these intracellular compartments, Lck signals for Ras and Erk activation, which triggers IL-2 production. IL-2 causes T cells to grow and proliferate, leading to more T cells that HIV can infect and leading to activation of the machinery HIV needs to replicate itself within infected T cells. Nef triggers lysosomal degradation of CTLA-4. This is because CTLA-4 can serve as an off-switch for T cells, which would lead to inhibition of HIV replication if active. Nef inactivates the Bad protein via phosphorylation. Bad participates in apoptotic cascades, so Nef prevents apoptosis of the infected host cell in this way.

Vif forms a complex with the host antiviral proteins APOBEC3F and APOBEC3G and induces their ubiquitination and subsequent degradation by the proteosome.16 It also may inhibit these proteins through other mechanisms. APOBEC3F and APOBEC3G are cytidine deaminases that hypermutate the negative-sense strand of HIV cDNA, leading to weak or nonviable viruses.17 These proteins also interfere with reverse transcription by blocking tRNALys3 from binding to the HIV RNA 5’UTR (tRNALys3 usually acts as a primer to initiate reverse transcription of the HIV genome).18

Vpr facilitates nuclear import of the HIV pre-integration complex.19 The pre-integration complex consists of viral cDNA and associated proteins (uncoating and reverse transcription have already occurred at this stage). Vpr binds the pre-integration complex and recruits host importins to enable nuclear import. It may further enhance nuclear import through interactions with some of the nuclear pore proteins. In addition to nuclear import, Vpr has several more functions: it acts as a coactivator (along with other proteins) of the HIV 5’UTR’s U3 promoter, might influence NF-κB regulation, may modulate apoptotic pathways, and arrests the cell cycle at the G2 stage.

Life cycle:

CD4+ T cells represent the primary targets of HIV, though the virus is also capable of infecting other cell types such as dendritic cells.20 HIV infects CD4+ T cells through binding its gp120 glycoprotein to the CD4 receptor and the CCR5 coreceptor or the CXCR4 coreceptor.10 This triggers fusion of the viral envelope with the plasma membrane and allows the core capsid to enter the cytosol.

HIV’s core capsid is transported by motor proteins along microtubules to dock at nuclear pores. The nuclear pore complex has flexible cytosolic filaments composed primarily of the Nup358 protein, which interacts with the core capsid.21 These interactions guide the narrow end of the core capsid into the nuclear pore’s central channel. Next, the core capsid interacts with the central channel’s unstructured phenylalanine-glycine (FG) repeats that exist in a hydrogel-like liquid phase. As the core capsid translocates through the central pore, it binds the Nup153 protein, a component of the nuclear pore complex’s basket. Finally, many copies of the nucleoplasmic CPSF6 protein coat the core capsid and escort it towards its genomic site of integration. It is thought that the reverse transcription process begins inside of the core capsid at this point, leading to cDNA synthesis.21,22 Buildup of newly made cDNA within the core capsid likely results in pressure that helps rupture the capsid structure, facilitating uncoating.

Tetrameric HIV integrase binds both of the viral LTRs and facilitates integration of the cDNA into the host genome.23 Though integration sites vary widely, they are not entirely random. Host chromatin structure and other factors influence where the viral cDNA integrates.24 Transcription of HIV RNAs can then proceed from the U3 promoter with the aid of the Tat protein and host factors. As described earlier, a series of RNA splicing events produce the various RNAs necessary to synthesize all of the different HIV proteins and polyproteins.

Env protein is trafficked to the cell membrane through the secretory pathway. It is cleaved by a host furin enzyme into gp41 and gp160 components during its time in the endoplasmic reticulum.9 Gag and Gag-Pol polyproteins are expressed cytosolically. Since Gag is post-translationally modified by amino-terminal myristoylation, it anchors to the cell membrane by inserting its myristate tail into the lipid bilayer.25 Gag and a smaller number of Gag-Pol accumulate on the inner membrane surface and incorporate gp41-gp160 complexes. NC domains in the Gag proteins bind and help package the two copies of HIV genomic RNA. The p6 region of the Gag protein (located at the C-terminal end) then recruits host ESCRT-I and ALIX proteins, which subsequently sequester host ESCRT-III and VPS4 complexes to drive budding and membrane scission, releasing virus into the extracellular space. After this, the HIV viral protease (from within the Gag-Pol polyprotein) cleaves the Gag and Gag-Pol polyproteins into their constituent proteins, facilitating maturation of the released HIV particles.

SARS-CoV-2

Genome and Structure:

The SARS-CoV-2 genome consists of about 30 kb of linear positive-sense ssRNA. There is a m7G-cap (specifically m7GpppA1) at the 5’ end of the genome and a 30-60 nucleotide poly-A tail at the 3’ end of the genome. These protective structures minimize exonuclease degradation.26 The genome also has a 5’ UTR and a 3’ UTR which contain sequences that aid in transcriptional regulation and in packaging. The SARS-CoV-2 genome directly translates two partially overlapping polyproteins, ORF1a and ORF1b. There is a –1 ribosomal frameshift in ORF1b relative to ORF1a. Within the polyproteins, two self-activating proteases (Papain-like protease PLpro and 3-chymotrypsin-like protease 3CLpro) perform cleavage events that lead to the generation of the virus’s 16 non-structural proteins (nsps). It should be noted that the 3CLpro is also known as the main protease or Mpro. The coronavirus also produces 4 structural proteins, but these are not translated until after the synthesis of corresponding subgenomic RNAs via the viral replication complex. To create these subgenomic RNAs, negative-sense RNA must first be made and then undergo conversion back to positive-sense RNA for translation. Genes encoding the structural proteins are located downstream of the ORF1b section.

SARS-CoV-2’s four structural proteins include the N, E, M, and S proteins. Many copies of the N (nucleocapsid) protein bind the RNA genome and organize it into a helical ribonucelocapsid complex. The complex undergoes packaging into the viral envelope during coronavirus budding. Interactions between the N protein and the other structural proteins may facilitate this packaging process. The N protein also inhibits host immune responses by antagonizing viral suppressor RNAi and by blocking the signaling of interferon production pathways.27

The transmembrane E (envelope) protein forms pentamers and plays a key but poorly understood role in the budding of viral envelopes into the endoplasmic reticulum Golgi intermediate compartment (ERGIC).28–30 Despite its importance in budding, mature viral particles do not incorporate very many E proteins into their envelopes. One of the posttranslational modifications of the E protein is palmitoylation. This aids subcellular trafficking and interactions with membranes. E protein pentamers also act as ion channels that alter membrane potential.31,32 This may lead to inflammasome activation, a contributing factor to cytokine storm induction.

The M (membrane) protein is the most abundant protein in the virion and drives global curvature in the ERGIC membrane to facilitate budding.30,33 It forms transmembrane dimers that likely oligomerize to induce this curvature.34 The M protein also has a cytosolic (and later intravirion) globular domain that likely interacts with the other structural proteins. M protein dimers also induce local curvature through preferential interactions with phosphatidylserine and phosphatidylinositol lipids.29,30 M proteins help sequester S proteins into the envelopes of budding viruses.35

The S (spike) protein of SARS-CoV-2 has been heavily studied due to its central roles in the infectivity and immunogenicity of the coronavirus. It forms a homotrimer that protrudes from the viral envelope and is heavily glycosylated. It binds the host’s ACE2 receptor (angiotensin-converting enzyme 2 receptor) and undergoes conformational changes to promote viral fusion.36 The S protein undergoes cleavage into S1 and S2 subunits by the host’s furin protease during viral maturation.37,38 This enhances SARS-CoV-2 entry into lung cells and may partially explain the virus’s high degree of transmissibility. The S1 fragment contains the receptor binding domain (RBD) and associated machinery while the S2 fragment facilitates fusion. Prior to cellular infection, most S proteins exist in a closed prefusion conformation where the RBDs of each monomer are hidden most of the time.39 After the S protein binds ACE2 during transient exposure of one of its RBDs, the other two RBDs quickly bind as well. This binding triggers a conformational change in the S protein that loosens the structure, unleashing the S2 fusion component and exposing another proteolytic cleavage site called S2’. Host transmembrane proteases such as TMPRSS2 cut at S2’, causing the full activation of the S2 fusion subunit and the dramatic elongation of the S protein into the postfusion conformation. This results in the viral envelope fusing with the host membrane and uptake of the coronavirus’s RNA into the cell.

The 16 nsps of SARS-CoV-2 play a variety of roles. For instance, nsp1 shuts down host cell translation by plugging the mRNA entry channel of the ribosome, inhibiting the host cell’s immune responses and maximizing viral production.40,41 Viral proteins still undergo translation because a conserved sequence in the coronavirus RNA helps circumvent the blockage through a poorly understood mechanism. The nsp5 protein is the protease 3CLpro.42 The nsp3 protein contains several subcomponents, including the protease PLpro. The nsp12, nsp7, and nsp8 proteins come together to form the RNA-dependent RNA polymerase (RdRp) that replicates the viral genome.42,43 The nsp2 protein is likely a topoisomerase which functions in RNA replication. The nsp4 and nsp6 proteins as well as certain subcomponents of nsp3 restructure intracellular host membranes into double-membrane vesicles (DMVs) which compartmentalize viral replication.44

Beyond the 4 structural proteins and 16 nsps of SARS-CoV-2, the coronaviral genome also encodes some poorly understood accessory proteins including ORF3a, ORF3b, ORF6, ORF7a, ORF7b, ORF8 and ORF9b.45 These accessory proteins are non-essential for replication in vitro, but they are thought to be required for the virus’s full degree of virulence in vivo.

Life cycle:

As mentioned, SARS-CoV-2 infects cells by first binding a S protein RBD to the ACE2 receptor. This triggers a conformational change that elongates the S protein’s structure and reveals the S2 fusion fragment, facilitating fusion of the virion envelope with the host cell membrane.39 Cleavage of the S’ site by proteases like TMPRSS2 aid this change from the prefusion to postfusion configurations. Alternatively, SARS-CoV-2 can enter the cell by binding to ACE2, undergoing endocytosis, and fusing with the endosome to release its genome (as induced by endosomal cathepsin proteases).45 After release of the SARS-CoV-2 genome into the cytosol, the N protein disassociates and allows translation of ORF1a and ORF1b, producing polyproteins which are cleaved into mature proteins by the PLpro and 3CLpro proteases as discussed earlier. 

The RdRp complex synthesizes negative-sense full genomic RNAs as well as negative-sense subgenomic RNAs. In the latter case, discontinuous transcription is employed, a process by which the RdRp jumps over certain sections of the RNA and initiates transcription separately from the rest of the genome.46 The negative-sense RNAs are subsequently converted back into positive-sense full genomic RNAs and positive-sense subgenomic RNAs. The subgenomic RNAs are translated to make structural proteins and some accessory proteins.45

As described earlier, the nsp4, nsp6, and parts of nsp3 proteins remodel host endoplasmic reticulum (ER) to create DMVs.45 These DMVs are the site of the coronaviral genomic replication and serve to shield the viral RNA and RdRp complex from cellular innate immune factors. DMVs cluster together and are continuous with the ER mostly through small tubular connections. After replication, the newly synthesized coronavirus RNAs undergo export into the cytosol through molecular pore complexes that span both membranes of the DMVs.47 These molecular pore complexes are composed of nsp3 domains and possibly other viral and/or host proteins.

Newly replicated SARS-CoV-2 genomic RNAs complex with N proteins to form helical nucleocapsids. To enable packaging, the nucleocapsids interact with M protein cytosolic domains which protrude at the ERGIC.48 M proteins, E proteins, and S proteins are all localized to the ERGIC membrane. The highly abundant M proteins induce curvature of the membrane to facilitate budding. As mentioned, E proteins also play essential roles in budding, but the mechanisms are poorly understood. Once the virions have budded into the ERGIC, they are shuttled through the Golgi via a series of vesicles and eventually secreted out of the cell.

Adeno-associated virus (AAV)

Genome and Structure:

AAV genomes are about 4.7 kb in length and are composed of ssDNA. Inverted terminal repeats (ITRs) form hairpin structures at ends of the genome. These ITR structures are important for AAV genomic packaging and replication. Rep genes (encoded via overlapping reading frames) include Rep78, Rep68, Rep52, Rep40.49 These proteins facilitate replication of the viral genome. As a Dependoparvovirus, additional helper functions from adenovirus (or certain other viruses) are needed for AAVs to replicate.

AAV capsids are about 25 nm in diameter. Cap genes include VP1, VP2, VP3 and are transcribed from overlapping reading frames.50 The VP3 protein is the smallest capsid protein. The VP2 protein is the same as VP3 except that it includes an N-terminal extension with a nuclear localization sequence. The VP1 protein is the same as VP2 except that it includes a further N-terminal extension encoding a phospholipase A2 (PLA2) that facilitates endosomal escape during infection. In the AAV capsid, VP1, VP2, and VP3 are present at a ratio of roughly 1:1:10. It should be noted that this ratio is actually the average of a distribution, not a fixed number.

Frame-shifted start codons in the Cap gene region transcribe AAP (assembly activating protein) and MAAP (membrane associated accessory protein). These proteins help facilitate packaging and other aspects of the AAV life cycle.

Life cycle:

There are a variety of different AAV serotypes (AAV2, AAV6, AAV9, etc.) that selectively infect certain tissue types. AAVs bind to host cell receptors and are internalized by endocytosis. The particular receptors involved can vary depending on the AAV serotype, though some receptors are consistent across many serotypes. Internalization occurs most often via clathrin-coated pits, but some AAVs are internalized by other routes such as macropinocytosis or the CLIC/GEEC tubulovesicular pathway.51

After endocytosis, conformational changes in the AAV capsid lead to exposure of the PLA2 VP1 domain, which facilitates endosomal escape. The AAV is then transported to the nucleus mainly by motor proteins on cytoskeletal highways. It enters via nuclear pores and finishes uncoating its genome.

AAV genomes initiate replication using the ends of their ITR hairpins as primers. This leads to a series of complex steps involving strand displacement and nicking.49 In the end, new copies of the AAV genome are synthesized. The Rep proteins are key players in this process. It is important to realize that AAVs can only replicate in cells which have also been infected by adenovirus or similar helper viruses (this is why they are called “adeno-associated viruses”). Adenoviruses provide helper genes encoding proteins (e.g. E4, E2a, VA) that are vital for the successful completion of the AAV life cycle. After new AAV capsids have assembled from VP1, VP2, and VP3 and once AAV genomes have been replicated, the ssDNA genomes are threaded into the capsids via pores at their five-fold vertices.

AAVs are nonpathogenic, though a large fraction of people possess antibodies against at least some serotypes, so exposure to them is fairly common.

Adenovirus

Genome and Structure:

Adenovirus genomes are about 36 kb in size and are composed of linear dsDNA. They possess inverted terminal repeats (ITRs) which help facilitate replication and other functions. These genomes contain a variety of transcriptional units which are expressed at different times during the virus’s life cycle.52 E1A, E1B, E2A, E2B, E3, and E4 transcriptional units are expressed early during cellular infection. Their proteins are involved in DNA replication, transcriptional regulation, and suppression of host immune responses. The L1, L2, L3, L4, and L5 transcriptional units are expressed later in the life cycle. Their products include most of the capsid proteins as well as other proteins involved in packaging and assembly. Each transcriptional unit can produce multiple mRNAs through the host’s alternative splicing machinery.

The capsid of the adenovirus is about 90 nm in diameter and consists of three major proteins (hexon, penton, and fiber proteins) as well as a variety of minor proteins and core proteins. Hexon trimer is the most abundant protein in the capsid, the pentameric pentons occur at the vertices, and trimeric fibers are positioned on top of the pentons.53 The fibers point outwards from the capsid and end in knob domains which bind to cellular receptors. In Ad5, a commonly studied type of adenovirus, the fiber knob primarily binds to the coxsackievirus and adenovirus receptor (CAR). That said, it should be noted that Ad5’s fiber knob can also bind to alternative receptors such as vascular cell adhesion molecule 1 and heparan sulfate proteoglycans.

Minor capsid proteins include pIX, pIIIa, pVI, and pVIII. The pIX protein interlaces between hexons and helps stabilize the capsid. Though pIX is positioned in the crevices between the hexons, it is still exposed to the outside environment. By contrast, the pIIIa, pVI, and pVIII proteins bind to the inside of the capsid and contribute further structural stabilization. When the adenovirus is inside of the acidic endosome during infection, conformational changes in the capsid release the pVI protein, which facilitates endosomal escape through membrane lytic activity.

Adenovirus core proteins include pV, pVII, protein μ (also known as pX), adenovirus proteinase (AVP), pIVa2, and terminal protein (TP).54 The pVII protein has many positively-charged arginine residues and so functions to condense the viral DNA. The pV protein bridges the core with the capsid through interactions with pVII and with pVI. AVP cleaves various adenoviral proteins (pIIIa, TP, pVI, pVII, pVIII, pX) to convert them to their mature forms.55 The pIVa2 and pX proteins interact with the viral DNA and may play roles in packaging or replication. TP binds to the ends of the genome and is essential for localizing the viral DNA in the nucleus and for viral replication.

Life Cycle:

Adenovirus infects cells by binding its fiber knob to cellular receptors such as CAR (in the case of Ad5). The penton then binds certain αv integrins, positioning the viral capsid for endocytosis.56 When the endosome acidifies, the adenovirus capsid partially disassembles, fibers and pentons fall away, and pVI is released.57 The pVI protein’s membrane lytic activity facilitates endosomal escape. Partially disassembled capsids then undergo dynein-mediated transport along microtubules and dock at the entrance to nuclear pores. The capsids further disassemble and releases DNA through the nuclear pore. This DNA remains complexed with pVII after it enters the nucleus.

Adenoviral transcription is initiated by the E1A protein, inducing expression of early genes.58 This subsequently leads to expression of the E2, E3, and E4 transcriptional units, which help the virus escape immune responses. This cascade leads to expression of the L1, L2, L3, L4, and L5 transcriptional units, which mainly synthesize viral structural proteins and facilitate capsid assembly.

In the nucleus, adenovirus genomes replicate within dense complexes of protein that can be seen as spots via fluorescence microscopy. Replication begins at the ITRs and is primed by TP.59 Several more viral proteins and host proteins also aid the initiation of replication. Nontemplate strands are displaced during replication but may reanneal and act as template strands later. Adenovirus DNA binding protein and adenovirus DNA polymerase play important roles in replication. Once the genome has been replicated, TP undergoes cleavage into its mature form, signaling for packaging of new genomes.

The adenoviral capsid assembly and maturation process occurs in the nucleus.58 Once enough assembled adenoviruses have accumulated, they rupture the nuclear membrane using adenoviral death protein and subsequently lyse the cell, releasing adenoviral particles.

Herpes Simplex Virus 1 (HSV-1)

Genome and Structure:

HSV-1 genomes are about 150 kb in size and are composed of linear dsDNA. These genomes include a unique long (UL) region and a unique short (US) region.60 The UL and US regions are both flanked by their own inverted repeats. The terminal inverted repeats are called TRL and TRS while the internal inverted repeats are called IRL and IRS. HSV-1 contains approximately 80 genes, though the complexity of its genomic organization makes an exact number of genes difficult to obtain. As with many other viruses, HSV-1 genomes encode early, middle, and late genes. The early genes activate and regulate transcription of the middle and late genes. Middle genes facilitate genome replication and late genes mostly encode structural proteins.

The diameter of HSV-1 ranges around 155 nm to 240 nm.61 Its virions include an inner icosahedral capsid (with a 125 nm diameter) surrounded by tegument proteins which are in turn enveloped by a lipid membrane containing glycoproteins.

HSV-1’s icosahedral capsid consists of a variety of proteins. Some of the most important capsid proteins are encoded by the UL19, UL18, UL38, UL6, UL17, and UL25 genes.62 The UL19 gene encodes the major capsid protein VP5, which forms pentamers and hexamers for the capsid. These VP5 pentamers and hexamers are glued together by triplexes consisting of two copies of VP23 (encoded by UL18) and one copy of VP19C (encoded by UL38).63 The UL6 gene encodes the protein that makes up the portal complex, a structure used by HSV-1 to release its DNA during infection. Each HSV-1 capsid has a single portal (composed of 12 copies of the portal protein) located at one of the vertices. UL17 and UL25 encode additional structural proteins that stabilize the capsid by binding on top of the other vertices. These two proteins also serve as a bridge between the capsid core and the tegument proteins.

The tegument of HSV-1 contains dozens of distinct proteins. Some examples include pUL36, pUL37, pUL7, and pUL51 proteins. The major tegument proteins are pUL36 and pUL37. The pUL36 protein binds on top of the UL17-UL25 complexes at the capsid’s vertices.64 The pUL37 protein subsequently associates with pUL36. The pUL51 protein associates with cytoplasmic membranes in infected cells and recruits the pUL7 protein.65 This pUL51-pUL7 interaction is important for HSV-1 assembly. HSV-1 has many more tegument proteins which play various functional roles.

HSV-1’s envelope contains up to 16 unique glycoproteins. Four of these glycoproteins (gB, gD, gH, and gL) are essential for viral entry into cells.66 The gD glycoprotein first binds to one of its cellular receptors (nectin-1, herpesvirus entry mediator or HVEM, or 3-O-sulfated heparan sulfate). This binding event triggers a conformational change in gD that allows it to activate the gH/gL heterodimer. Next, gH/gL activate gB which induces fusion of HSV-1’s envelope with the cell membrane. Though the remaining 12 envelope glycoproteins are poorly understood, it is thought that they also play roles that influence cellular tropism and entry.

Life cycle:

After binding to cellular receptors via its glycoproteins, HSV-1 induces fusion of its envelope with the host cell membrane.67 The capsid is trafficked to nuclear pores via microtubules. Since the capsid is too large to pass through a nuclear pore directly, the virus instead ejects its DNA through the pore via the portal complex.68

HSV-1 replicates its genome and assembles its capsids in the nucleus. But the assembled capsids are again too large to exist the nucleus through nuclear pores. To overcome this issue, HSV-1 first buds via the inner nuclear membrane into the perinuclear cleft (the space between nuclear membranes), acquiring a primary envelope.67 This process is driven by a pair of proteins (pUL34 and pUL31) which together form the nuclear egress complex. Next, the primary envelope fuses with the outer nuclear membrane, releasing the assembled capsids into the cytosol.

To acquire its final envelope, the HSV-1 capsid likely buds into the trans-Golgi network or into certain tubular vesicular organelles.69 These membrane sources contain the envelope proteins of the virus as produced by transcription and various secretory pathways. One player is the pUL51 tegument protein that starts associated with the membrane into which the virus buds. The interaction between pUL51 and pUL7 helps facilitate recruitment of the capsid to the membrane. (Capsid envelopment is also coupled in many other ways to formation of the outer tegument). The enveloped virion eventually undergoes trafficking through the secretory system and eventually is packaged into exosomes that fuse with the cell membrane and release completed virions into the extracellular environment.

In humans, HSV-1 infects the epithelial cells first and produces viral particles.70 It subsequently enters the termini of sensory neurons, undergoes retrograde transport into the brain, and remains in the central nervous system in a dormant state. During periods of stress in the host, the virus is reactivated and undergoes anterograde transport to infect epithelial cells once again.

References

1.        Wain-Hobson, S., Sonigo, P., Danos, O., Cole, S. & Alizon, M. Nucleotide sequence of the AIDS virus, LAV. Cell 40, 9–17 (1985).

2.        Wilusz, J. Putting an ‘End’ to HIV mRNAs: capping and polyadenylation as potential therapeutic targets. AIDS Res. Ther. 10, 31 (2013).

3.        Marcello, A., Zoppé, M. & Giacca, M. Multiple Modes of Transcriptional Regulation by the HIV-1 Tat Transactivator. IUBMB Life 51, 175–181 (2001).

4.        Brigati, C., Giacca, M., Noonan, D. M. & Albini, A. HIV Tat, its TARgets and the control of viral gene expression. FEMS Microbiol. Lett. 220, 57–65 (2003).

5.        Harrison, J. J. E. K. et al. Cryo-EM structure of the HIV-1 Pol polyprotein provides insights into virion maturation. Sci. Adv. 8, eabn9874 (2022).

6.        Guerrero, S. et al. HIV-1 Replication and the Cellular Eukaryotic Translation Apparatus. Viruses vol. 7 199–218 at https://doi.org/10.3390/v7010199 (2015).

7.        Feinberg, M. B. & Greene, W. C. Molecular insights into human immunodeficiency virus type 1 pathogenesis. Curr. Opin. Immunol. 4, 466–474 (1992).

8.        Sertznig, H., Hillebrand, F., Erkelenz, S., Schaal, H. & Widera, M. Behind the scenes of HIV-1 replication: Alternative splicing as the dependency factor on the quiet. Virology 516, 176–188 (2018).

9.        Behrens, A.-J. & Crispin, M. Structural principles controlling HIV envelope glycosylation. Curr. Opin. Struct. Biol. 44, 125–133 (2017).

10.      Campbell, E. M. & Hope, T. J. HIV-1 capsid: the multifaceted key player in HIV-1 infection. Nat. Rev. Microbiol. 13, 471–483 (2015).

11.      Andrew, A. & Strebel, K. HIV-1 Vpu targets cell surface markers CD4 and BST-2 through distinct mechanisms. Mol. Aspects Med. 31, 407–417 (2010).

12.      Bour, S., Geleziunas, R. & Wainberg, M. A. The human immunodeficiency virus type 1 (HIV-1) CD4 receptor and its central role in promotion of HIV-1 infection. Microbiol. Rev. 59, 63–93 (1995).

13.      Engelman, A. & Cherepanov, P. The structural biology of HIV-1: mechanistic and therapeutic insights. Nat. Rev. Microbiol. 10, 279–290 (2012).

14.      Marino, J., Wigdahl, B. & Nonnemacher, M. R. Extracellular HIV-1 Tat Mediates Increased Glutamate in the CNS Leading to Onset of Senescence and Progression of HAND   . Frontiers in Aging Neuroscience   vol. 12 at https://www.frontiersin.org/articles/10.3389/fnagi.2020.00168 (2020).

15.      Abraham, L. & Fackler, O. T. HIV-1 Nef: a multifaceted modulator of T cell receptor signaling. Cell Commun. Signal. 10, 39 (2012).

16.      Mehle, A. et al. Vif Overcomes the Innate Antiviral Activity of APOBEC3G by Promoting Its Degradation in the Ubiquitin-Proteasome Pathway *. J. Biol. Chem. 279, 7792–7798 (2004).

17.      Donahue, J. P., Vetter, M. L., Mukhtar, N. A. & D’Aquila, R. T. The HIV-1 Vif PPLP motif is necessary for human APOBEC3G binding and degradation. Virology 377, 49–53 (2008).

18.      Fei, G., Shan, C., Meijuan, N., Jenan, S. & Lawrence, K. Inhibition of tRNALys3-Primed Reverse Transcription by Human APOBEC3G during Human Immunodeficiency Virus Type 1 Replication. J. Virol. 80, 11710–11722 (2006).

19.      Kogan, M. & Rappaport, J. HIV-1 Accessory Protein Vpr: Relevance in the pathogenesis of HIV and potential for therapeutic intervention. Retrovirology 8, 25 (2011).

20.      Hladik, F. & McElrath, M. J. Setting the stage: host invasion by HIV. Nat. Rev. Immunol. 8, 447–457 (2008).

21.      Müller, T. G., Zila, V., Müller, B. & Kräusslich, H.-G. Nuclear Capsid Uncoating and Reverse Transcription of HIV-1. Annu. Rev. Virol. 9, 261–284 (2022).

22.      Müller, T. G. et al. HIV-1 uncoating by release of viral cDNA from capsid-like structures in the nucleus of infected cells. Elife 10, e64776 (2021).

23.      Marchand, C., Johnson, A. A., Semenova, E. & Pommier, Y. Mechanisms and inhibition of HIV integration. Drug Discov. Today Dis. Mech. 3, 253–260 (2006).

24.      Hughes, S. H. & Coffin, J. M. What Integration Sites Tell Us about HIV Persistence. Cell Host Microbe 19, 588–598 (2016).

25.      Freed, E. O. HIV-1 assembly, release and maturation. Nat. Rev. Microbiol. 13, 484–496 (2015).

26.      Brant, A. C., Tian, W., Majerciak, V., Yang, W. & Zheng, Z.-M. SARS-CoV-2: from its discovery to genome structure, transcription, and replication. Cell Biosci. 11, 136 (2021).

27.      Bai, Z., Cao, Y., Liu, W. & Li, J. The SARS-CoV-2 Nucleocapsid Protein and Its Role in Viral Structure, Biological Functions, and a Potential Target for Drug or Vaccine Mitigation. Viruses  vol. 13 at https://doi.org/10.3390/v13061115 (2021).

28.      Schoeman, D. & Fielding, B. C. Coronavirus envelope protein: current knowledge. Virol. J. 16, 69 (2019).

29.      Monje-Galvan, V. & Voth, G. A. Molecular interactions of the M and E integral membrane proteins of SARS-CoV-2. Faraday Discuss. (2021) doi:10.1039/D1FD00031D.

30.      Collins, L. T. et al. Elucidation of SARS-CoV-2 budding mechanisms through molecular dynamics simulations of M and E protein complexes. J. Phys. Chem. Lett. 12, 12249–12255 (2021).

31.      Arya, R. et al. Structural insights into SARS-CoV-2 proteins. J. Mol. Biol. 433, 166725 (2021).

32.      Yang, H. & Rao, Z. Structural biology of SARS-CoV-2 and implications for therapeutic development. Nat. Rev. Microbiol. 19, 685–700 (2021).

33.      J Alsaadi, E. A. & Jones, I. M. Membrane binding proteins of coronaviruses. Future Virol. 14, 275–286 (2019).

34.      Neuman, B. W. et al. A structural analysis of M protein in coronavirus assembly and morphology. J. Struct. Biol. 174, 11–22 (2011).

35.      Boson, B. et al. The SARS-CoV-2 envelope and membrane proteins modulate maturation and retention of the spike protein, allowing assembly of virus-like particles. J. Biol. Chem. 296, (2021).

36.      Zhang, J., Xiao, T., Cai, Y. & Chen, B. Structure of SARS-CoV-2 spike protein. Curr. Opin. Virol. 50, 173–182 (2021).

37.      Walls, A. C. et al. Structure, Function, and Antigenicity of the SARS-CoV-2 Spike Glycoprotein. Cell 181, 281-292.e6 (2020).

38.      Peacock, T. P. et al. The furin cleavage site in the SARS-CoV-2 spike protein is required for transmission in ferrets. Nat. Microbiol. 6, 899–909 (2021).

39.      Fertig, T. E. et al. The atomic portrait of SARS-CoV-2 as captured by cryo-electron microscopy. J. Cell. Mol. Med. 26, 25–34 (2022).

40.      Schubert, K. et al. SARS-CoV-2 Nsp1 binds the ribosomal mRNA channel to inhibit translation. Nat. Struct. Mol. Biol. 27, 959–966 (2020).

41.      Yuan, S. et al. Nonstructural Protein 1 of SARS-CoV-2 Is a Potent Pathogenicity Factor Redirecting Host Protein Synthesis Machinery toward Viral RNA. Mol. Cell 80, 1055-1066.e6 (2020).

42.      Raj, R. Analysis of non-structural proteins, NSPs of SARS-CoV-2 as targets for computational drug designing. Biochem. Biophys. Reports 25, 100847 (2021).

43.      Kirchdoerfer, R. N. & Ward, A. B. Structure of the SARS-CoV nsp12 polymerase bound to nsp7 and nsp8 co-factors. Nat. Commun. 10, 2342 (2019).

44.      Roingeard, P. et al. The double-membrane vesicle (DMV): a virus-induced organelle dedicated to the replication of SARS-CoV-2 and other positive-sense single-stranded RNA viruses. Cell. Mol. Life Sci. 79, 425 (2022).

45.      Baggen, J., Vanstreels, E., Jansen, S. & Daelemans, D. Cellular host factors for SARS-CoV-2 infection. Nat. Microbiol. 6, 1219–1232 (2021).

46.      Sashittal, P., Zhang, C., Peng, J. & El-Kebir, M. Jumper enables discontinuous transcript assembly in coronaviruses. Nat. Commun. 12, 6728 (2021).

47.      Wolff, G. et al. A molecular pore spans the double membrane of the coronavirus replication organelle. Science (80-. ). 369, 1395–1398 (2020).

48.      David, B. & Delphine, M. Betacoronavirus Assembly: Clues and Perspectives for Elucidating SARS-CoV-2 Particle Formation and Egress. MBio 12, e02371-21 (2021).

49.      Sha, S. et al. Cellular pathways of recombinant adeno-associated virus production for gene therapy. Biotechnol. Adv. 49, 107764 (2021).

50.      Wang, D., Tai, P. W. L. & Gao, G. Adeno-associated virus vector as a platform for gene therapy delivery. Nat. Rev. Drug Discov. 18, 358–378 (2019).

51.      Riyad, J. M. & Weber, T. Intracellular trafficking of adeno-associated virus (AAV) vectors: challenges and future directions. Gene Ther. 28, 683–696 (2021).

52.      Ahi, Y. S. & Mittal, S. K. Components of Adenovirus Genome Packaging. Frontiers in Microbiology vol. 7 1503 at https://www.frontiersin.org/article/10.3389/fmicb.2016.01503 (2016).

53.      Gallardo, J., Pérez-Illana, M., Martín-González, N. & San Martín, C. Adenovirus Structure: What Is New? International Journal of Molecular Sciences  vol. 22 at https://doi.org/10.3390/ijms22105240 (2021).

54.      Kulanayake, S. & Tikoo, S. K. Adenovirus Core Proteins: Structure and Function. Viruses  vol. 13 at https://doi.org/10.3390/v13030388 (2021).

55.      Russell, W. C. & Kemp, G. D. Role of Adenovirus Structural Components in the Regulation of Adenovirus Infection BT  – The Molecular Repertoire of Adenoviruses I: Virion Structure and Infection. in (eds. Doerfler, W. & Böhm, P.) 81–98 (Springer Berlin Heidelberg, 1995). doi:10.1007/978-3-642-79496-4_6.

56.      R., N. G. & L., S. P. Role of αv Integrins in Adenovirus Cell Entry and Gene Delivery. Microbiol. Mol. Biol. Rev. 63, 725–734 (1999).

57.      Pied, N. & Wodrich, H. Imaging the adenovirus infection cycle. FEBS Lett. 593, 3419–3448 (2019).

58.      Georgi, F. & Greber, U. F. The Adenovirus Death Protein – a small membrane protein controls cell lysis and disease. FEBS Lett. 594, 1861–1878 (2020).

59.      Hoeben, R. C. & Uil, T. G. Adenovirus DNA Replication. Cold Spring Harb. Perspect. Biol.  5, (2013).

60.      McGeoch, D. J., Rixon, F. J. & Davison, A. J. Topics in herpesvirus genomics and evolution. Virus Res. 117, 90–104 (2006).

61.      Laine, R. F. et al. Structural analysis of herpes simplex virus by optical super-resolution imaging. Nat. Commun. 6, 5980 (2015).

62.      Mettenleiter, T. C., Klupp, B. G. & Granzow, H. Herpesvirus assembly: a tale of two membranes. Curr. Opin. Microbiol. 9, 423–429 (2006).

63.      E., H. E. Up close with herpesviruses. Science (80-. ). 360, 34–35 (2018).

64.      H., F. W. et al. The Large Tegument Protein pUL36 Is Essential for Formation of the Capsid Vertex-Specific Component at the Capsid-Tegument Interface of Herpes Simplex Virus 1. J. Virol. 89, 1502–1511 (2015).

65.      J., R. R., Rachel, F. & M., L. R. The Herpes Simplex Virus 1 UL51 Protein Interacts with the UL7 Protein and Plays a Role in Its Recruitment into the Virion. J. Virol. 89, 3112–3122 (2015).

66.      T., H. A., E., D. R., E., H. E. & Thomas, S. Contributions of the Four Essential Entry Glycoproteins to HSV-1 Tropism and the Selection of Entry Routes. MBio 12, e00143-21 (2021).

67.      Zeev-Ben-Mordehai, T., Hagen, C. & Grünewald, K. A cool hybrid approach to the herpesvirus ‘life’ cycle. Curr. Opin. Virol. 5, 42–49 (2014).

68.      Newcomb, W. W., Cockrell, S. K., Homa, F. L. & Brown, J. C. Polarized DNA Ejection from the Herpesvirus Capsid. J. Mol. Biol. 392, 885–894 (2009).

69.      Ahmad, I. & Wilson, D. W. HSV-1 Cytoplasmic Envelopment and Egress. International Journal of Molecular Sciences  vol. 21 at https://doi.org/10.3390/ijms21175969 (2020).

70.      Roizman, B. & Whitley, R. J. An Inquiry into the Molecular Basis of HSV Latency and Reactivation. Annu. Rev. Microbiol. 67, 355–374 (2013).