Tuesday, December 30, 2014

A Tribute to Science and Reason -2014

Any one reading the news media would think of 2014 as a terrible year with wars, violence and epidemics; media saturated with news of ignorant people killing and getting killed for artificial divisions based on race, religion and nationalities. However behind scene and quietly, the scientific and technological progress that defines the human spirit have been taking at an ever expanding pace, and 2014 was another banner year for science. This is a short list of some of the fascinating new developments in science, medicine and technology in 2014. With a new scientific paper being published every 5 seconds, this is clearly an extremely short and incomplete list, a tiny sampling of all the major discoveries made in 2014 and also colored by my personal choices; but is meant more as a small tribute to the countless scientists, engineers, doctors and dreamers bringing about positive changes to us all through science, logic and reason. 



Piggy-backing proteins ride white blood cells to destroy metastasizing cancer

Cornell biomedical engineers have discovered a new way to destroy metastasizing cancer cells traveling through the bloodstream by hitching cancer-killing proteins along for a ride on life-saving white blood cells.
“These circulating cancer cells are doomed,” said Michael King, Cornell professor of biomedical engineering and the study’s senior author.
“About 90 percent of cancer deaths are related to metastases, but now we’ve found a way to dispatch an army of killer white blood cells that cause apoptosis — the cancer cell’s own death — obliterating them from the bloodstream.
“When surrounded by these guys, it becomes nearly impossible for the cancer cell to escape.”
Metastasis is the spread of a cancer cells to other parts of the body. Surgery and radiation are effective at treating primary tumors, but difficulty in detecting metastatic cancer cells has made treatment of the spreading cancers problematic, say the scientists.
King and his colleagues injected human blood samples, and later mice, with two proteins: E-selectin (ES, an adhesive) and TRAIL (Tumor Necrosis Factor Related Apoptosis-Inducing Ligand).
The TRAIL protein joined with the E-selectin protein was able to stick to leukocytes (white blood cells) abundant in the bloodstream. When a cancer cell comes into contact with TRAIL, which is nearly unavoidable in the frenzied flow of blood, the cancer cell essentially kills itself.


A step towards simulating a worm brain in a computer


The OpenWorm Project — an open-source project dedicated to creating a virtual C. elegans nematode in a computer by reverse-engineering its biology—  has now developed software that replicates the worm’s muscle movement. One can explore that with the OpenWorm browser, or the iOS OpenWorm 3D Browser appThe ultimate scientific goal of OpenWorm: understanding how the worm brain works via a full  digital simulation. 

Growing human organs inside pigs in Japan


Meiji University professor Hiroshi Nagashima is creating chimeric pigs, which carry genetic material from two different species, BBC Newsreports. It starts off by making what Nagashima calls “a-pancreatic” embryos. Inside the white pig embryo, the gene that carries the instructions for developing the animal’s pancreas has been “switched off.”
The Japanese team then introduces stem cells from a black pig into the embryo. What they have discovered is that as the pig develops, it will be normal except for its pancreas, which will be genetically a black pig’s.
In a lab at Tokyo University, Professor Hiro Nakauchi is taking the next step. He takes skin cells from an adult brown rat. He then uses gene manipulation to change these adult skin cells into induced pluripotent stem cells (iPS) cells, which can develop into any part of the animal’s body.
Nakauchi has succeeded in using these iPS cells to grow a brown rat pancreas inside a white mouse. He is hoping to develop a technique to take skin cells from a human adult and change them in to iPS cells. Those iPS cells can then be injected into a pig embryo. The technique is eventually to lead to development of human organs inside pigs to be used for transplantation. 

Google’s smart contact lens project could allow diabetics to track glucose levels automatically

To help people with diabetes as they try to keep their blood sugar levels under control, Google is testing a smart contact lens designed to measure glucose levels in tears.

It uses a tiny wireless chip and miniaturized glucose sensor that are embedded between two layers of soft contact lens material, according to Google Official Blog.

People with diabetes must still prick their finger and test drops of blood throughout the day. It’s disruptive, and it’s painful. And, as a result, many people with diabetes check their blood glucose less often than they should. This should help.

“We’re testing prototypes that can generate a reading once per second. We’re also investigating the potential for this to serve as an early warning for the wearer, so we’re exploring integrating tiny LED lights that could light up to indicate that glucose levels have crossed above or below certain thresholds. It’s still early days for this technology, but we’ve completed multiple clinical research studies which are helping to refine our prototype. We hope this could someday lead to a new way for people with diabetes to manage their disease.”

Google plans to look for partners who are experts in bringing products like this to market and in developing apps that make the measurements available to the wearer and their doctor.

Turning off ageing genes

This research came from the labs of Dr. Yishak, a leader in the growing field of genome-scale metabolic modeling or GSMMs. Using mathematical equations and computers, GSMMs describe the metabolism, or life-sustaining, processes of living cells. His algorithm, which is called a “metabolic transformation algorithm,” or MTA, can take information about any two metabolic states and predict the environmental or genetic changes required to go from one state to the other.

“Gene expression” is the measurement of the expression level of individual genes in a cell, and genes can be “turned off” in various ways to prevent them from being expressed in the cell. In this study, researchers applied MTA to the genetics of aging. After using custom-designed MTA to confirm previous laboratory findings, he used it to predict genes that can be turned off to make the gene expression of old yeast look like that of young yeast. Yeast is the most widely used genetic model because much of its DNA is preserved in humans. Researchers there found that turning off two of the genes, GRE3 and ADH2, in actual (non-digital) yeast significantly extends the yeast’s lifespan.

“You would expect about three percent of yeast’s genes to be lifespan-extending,” said Yizhak. “So achieving a 10-fold increase over this expected frequency, as we did, is very encouraging.”

Self driving cars (SDC)

Self-driving cars (SDC) that include driver control are expected to hit highways around the globe before 2025 and self-driving “only” cars (only the car drives) are anticipated around 2030, according to an emerging technologies study on Autonomous Cars from IHS Automotive. The price premium for the SDC electronics technology will add between $7,000 and $10,000 to a car’s sticker price in 2025, a figure that will drop to around $5,000 in 2030 and about $3,000 in 2035 when no driver controls are available.


Ultra thin skin to help with continuous patient monitoring

An entirely new approach to measuring body temperature — an ”electronic skin” that adheres non-invasively to human skin, conforms well to contours, and provides a detailed temperature map of any surface of the body — has been developed by an international multidisciplinary team including researchers at the University of Illinois at Urbana/Champaign and the National Institute of Biomedical Imaging and Bioengineering (NIBIB).

Vapor nonbubbles rapidly detect malaria through the skin

Rice University researchers have developed a noninvasive technology that accurately detects even a single malaria-infected cell among a million normal cells through the skin in seconds with a laser scanner.

The “vapor nanobubble” technology requires no dyes or diagnostic chemicals, there is no need to draw blood, and there are zero false-positive readings. The diagnosis and screening will be supported by a low-cost, battery-powered portable device that can be operated by non-medical personnel. One device should be able to screen up to 200,000 people per year, with the cost of diagnosis estimated to be below 50 cents, the researchers say.

Discovery of quantum vibrations in microtubules inside brain neurons corroborates controversial 20-year-old theory of consciousness

A review and update of a controversial 20-year-old theory of consciousness published in  Elsevier’s Physics of Life Reviews (open access) claims that consciousness derives from deeper-level, finer-scale activities inside brain neurons.

The recent discovery of quantum vibrations in microtubules inside brain neurons corroborates this theory, according to review authors Stuart Hameroff and Sir Roger Penrose. They suggest that EEG rhythms (brain waves) also derive from deeper level microtubule vibrations, and that from a practical standpoint, treating brain microtubule vibrations could benefit a host of mental, neurological, and cognitive conditions. Microtubules are major components of the structural skeleton of cells.

The theory, called “orchestrated objective reduction” (“Orch OR”), was first put forward in the mid-1990s by eminent mathematical physicist Sir Roger Penrose, FRS, Mathematical Institute and Wadham College, University of Oxford, and prominent anesthesiologist Stuart Hameroff, MD, Anesthesiology, Psychology and Center for Consciousness Studies, The University of Arizona, Tucson.


World’s first $1,000 genome enables ‘factory’ scale sequencing for population and disease studies

Illumina, Inc. announced Tuesday that its new HiSeq X Ten Sequencing System has broken the “sound barrier” of human genomics by enabling the $1,000 genome.

“This platform includes dramatic technology breakthroughs that enable researchers to undertake studies of unprecedented scale by providing the throughput to sequence tens of thousands of human whole genomes in a single year in a single lab,” Illumina stated.

Initial customers for the HiSeq X Ten System, which will ship in Q1 2014, include Macrogen, based in Seoul, South Korea and its CLIA laboratory in Rockville, Maryland, the Broad Institute in Cambridge, Massachusetts, and the Garvan Institute of Medical Research in Sydney, Australia.

“For the first time, it looks like it will be possible to deliver the $1,000 genome, which is tremendously exciting,” said Eric Lander, founding director of the Broad Institute and a professor of biology at MIT. “The HiSeq X Ten should give us the ability to analyze complete genomic information from huge sample populations. Over the next few years, we have an opportunity to learn as much about the genetics of human disease as we have learned in the history of medicine.”

“The HiSeq X Ten is an ideal platform for scientists and institutions focused on the discovery of genotypic variation to enable a deeper understanding of human biology and genetic disease,” Illumina stated. “It can sequence tens of thousands of samples annually with high-quality, high-coverage sequencing, delivering a comprehensive catalog of human variation within and outside coding regions.”

HiSeq X Ten utilizes a number of advanced design features to generate massive throughput. Patterned flow cells, which contain billions of nanowells at fixed locations, combined with a new clustering chemistry deliver a significant increase in data density (6 billion clusters per run). Using state-of-the art optics and faster chemistry, HiSeq X Ten can process sequencing flow cells more quickly than ever before — generating a 10x increase in daily throughput when compared to current HiSeq 2500 performance.

The HiSeq X Ten is sold as a set of 10 or more ultra-high throughput sequencing systems, each generating up to 1.8 terabases (Tb) of sequencing data in less than three days or up to 600 gigabases (Gb) per day, per system, providing the throughput to sequence tens of thousands of high-quality, high-coverage genomes per year.

Ultrasound directed to the human brain can boost spatial resolution

Virginia Tech Carilion Research Institute scientists have found that ultrasound directed to a specific region of the brain can boost performance in sensory discrimination. The study provides the first demonstration that low-intensity, transcranial-focused ultrasound (tFUS) can modulate human brain activity to enhance perception.

“Ultrasound has great potential for bringing unprecedented resolution to the growing trend of mapping the human brain’s connectivity,” said William “Jamie” Tyler, an assistant professor at the Virginia Tech Carilion Research Institute, who led the study. “So we decided to look at the effects of ultrasound on the region of the brain responsible for processing tactile sensory inputs.”

How to monitor drug effects in real time

A device that can monitor the levels of specific drugs as they flow through the bloodstream may soon take the guesswork out of drug dosing and allow physicians to tailor prescriptions to their patients’ specific biology.

Developed by UC Santa Barbara researchers Tom Soh,Kevin Plaxco and Scott Ferguson, the biosensor combines  engineering and biochemistry and has far-reaching potential. Doctors and pharmaceutical companies can generally determine reasonable drug doses for most patients through batteries of tests and trials. However, the efficacy of a drug treatment relies on maintaining therapeutic levels of the drug in the body, a feat not so easily accomplished.

“Current dosing regimens are really quite primitive,” said Plaxco, professor of chemistry and of biomolecular science and engineering. They rely on a patient’s age or body weight and are unable to account for specific responses over time. Drug levels may be influenced by individual patients’ metabolisms, or even by the foods they eat or other drugs they might be taking.

When coupled with the primitive state of current dosing algorithms, this variability can be quite dangerous for drugs that have narrow therapeutic ranges. That in turn requires both doctor and patient to balance on the fine line between effectiveness and toxicity.

Scientists could be one giant step closer to dispelling the uncertainty around patients’ biological responses as they receive theseCalled MEDIC (Microfluidic Electrochemical Detector for In vivo Concentrations), the instrument can determine — continuously and in real time — concentrations of specific molecules in tiny amounts of whole blood. The kind of information it can provide could lead to truly personalized medicine.

“The easier and faster your doctor can detect specific molecules — drug molecules, proteins that are diagnostic of a specific disease — the faster your doctor can diagnose disease and monitor treatment,” said Plaxco.

“For the first time, we can see how the body is processing specific molecules,” said Ferguson, a postdoctoral researcher in the Soh lab and the lead author of the study. Ferguson invented the central technology for continuously monitoring drug levels in whole blood, an element of time that is necessary to provide a full picture of how an individual responds to a certain drug.


A Wikipedia for robots

European scientists from six institutes and two universities have developed an online platform where robots can learn new skills from each other worldwide — a kind of “Wikipedia for robots.” The objective is to help develop robots better at helping elders with caring and household tasks. “The problem right now is that robots are often developed specifically for one task”, says René van de Molengraft, TU/e researcher and RoboEarthproject leader.

RoboEarth simply lets robots learn new tasks and situations from each other. All their knowledge and experience are shared worldwide on a central, online database.”

In addition, some computing and “thinking” tasks can be carried out by the system’s “cloud engine,” he said, “so the robot doesn’t need to have as much computing or battery power on‑board.”

A less-expensive way to duplicate the complicated steps of photosynthesis in making fuel

Argonne National Laboratory researchers have found a new, more efficient, less-expensive way to make fuel — principally, hydrogen — from sunlight and water: linking a synthetic cobalt-containing catalyst to an organic light-sensitive molecule called a chromophore.

Chromophore molecules, such as chlorophyll, are involved in capturing light for photosynthesis. Currently, the most efficient methods we have for  making fuel involve rare and expensive metal catalysts, such as platinum. Although cobalt is significantly less efficient than platinum when it comes to light-induced hydrogen generation, the drastic price difference between the two metals makes cobalt the obvious choice as the foundation for a synthetic catalyst, said Argonne chemist Karen Mulfort.

The Argonne study wasn’t the first to look at cobalt as a potential catalytic material; however, a paper by the researchers in Physical Chemistry Chemical Physic identified a new mechanism to link the chromophore with the catalyst.

Previous experiments with cobalt attempted to connect the chromophore directly with the cobalt atom within the larger compound, but this eventually caused the hydrogen generation process to break down. Instead, the Argonne researchers connected the chromophore to part of a larger organic ring that surrounded the cobalt atom, which allowed the reaction to continue significantly longer.

Future studies in this arena could involve nickel- and iron-based catalysts — metals that are even more naturally abundant than cobalt, although they are not quite as effective natural catalysts. If additional research in this field eventually would bear fruit (pun intended!) then we would have the most green and abundant source of energy within human control - conversion of abundant solar energy to usable forms with the same technique plants have been using for billions of years.


New technique allows minimally invasive ‘nanobiopsies’ of living cells


Researchers at UC Santa Cruz (UCSC) have developed a robotic “nanobiopsy” system that can extract tiny samples from inside a living cell without killing it. The single-cell nanobiopsy technique is a powerful tool for scientists working to understand the dynamic processes that occur within living cells, according to Nader Pourmand, professor of biomolecular engineering in UCSC’s Baskin School of Engineering.

“We can take a biopsy from a living cell and go back to the same cell multiple times over a couple of days without killing it. With other technologies, you have to sacrifice a cell to analyze it,” said Pourmand, who leads the Biosensors and Bioelectrical Technology group at UCSC.

The nanobiopsy platform is the latest device his group has developed that uses nanopipettes, which are small glass tubes that taper to a fine tip with a diameter of just 50 to 100 nanometers. “We can create nanopipettes in the lab — it doesn’t require an expensive nanofabrication facility,” Pourmand said. “To go into a cell, however, the problem is that you cannot see the tip, even with a high-end microscope, so you don’t know how far away from the cell it is.”

Adam Seger, a postdoctoral researcher in the lab (now at MagArray in Sunnyvale), solved this problem by developing a feedback control system based on a customized scanning ion conductance microscope (SICM). The system uses an ion current across the tip of the nanopipette as a feedback signal, detecting a drop in the current when the tip gets close to the cell surface.

An automated control system positions the nanopipette tip just above the cell surface and then plunges it down quickly to penetrate the cell membrane. Manipulating the voltage triggers the controlled uptake of a minute quantity of cellular material. Combined with the next generation DNA sequencing which requires extremely small quantities of material for analysis, this approach will be a boon to various areas in Medicine, especially in cancer research and treatment,


Cosmic web imaged for the first time

Astronomers have discovered a distant quasar illuminating a vast nebula of diffuse gas, revealing, for the first time, part of the network of filaments thought to connect galaxies in a cosmic web. Researchers at the University of California, Santa Cruz led the study, published January 19 in NatureUsing the 10-meter Keck I Telescope at the W. M. Keck Observatory in Hawaii, the researchers detected a very large, luminous nebula of gas extending about 2 million light-years across intergalactic space.

“This is a very exceptional object: it’s huge, at least twice as large as any nebula detected before, and it extends well beyond the galactic environment of the quasar,” said first author Sebastiano Cantalupo, a postdoctoral fellow at UC Santa Cruz.

The standard cosmological model of structure formation in the universe predicts that galaxies are embedded in a cosmic web of matter, most of which (about 84 percent) is invisible dark matter. This web is seen in the results from computer simulations of the evolution of structure in the universe, which show the distribution of dark matter on large scales, including the dark matter halos in which galaxies form and the cosmic web of filaments that connect them. Gravity causes ordinary matter to follow the distribution of dark matter, so filaments of diffuse, ionized gas are expected to trace a pattern similar to that seen in dark matter simulations.

Until now, however, these filaments have never been seen. Intergalactic gas has been detected by its absorption of light from bright background sources, but those results don’t reveal how the gas is distributed. In this study, the researchers detected the fluorescent glow of hydrogen gas resulting from its illumination by intense radiation from the quasar.

“This quasar is illuminating diffuse gas on scales well beyond any we’ve seen before, giving us the first picture of extended gas between galaxies. It provides a terrific insight into the overall structure of our universe,” said coauthor J. Xavier Prochaska, professor of astronomy and astrophysics at UC Santa Cruz.

The hydrogen gas illuminated by the quasar emits ultraviolet light known as Lyman alpha radiation. The distance to the quasar is so great (about 10 billion light-years) that the emitted light is “stretched” by the expansion of the universe from an invisible ultraviolet wavelength to a visible shade of violet by the time it reaches the Keck Telescope.

Knowing the distance to the quasar, the researchers calculated the wavelength for Lyman alpha radiation from that distance (based on the doppler frequency shift — similar to the decreasing pitch of a train whistle as it travels away from you) and built a special filter for the telescope’s LRIS spectrometer to get an image at that wavelength.

“We have studied other quasars this way without detecting such extended gas,” Cantalupo said. “The light from the quasar is like a flashlight beam, and in this case we were lucky that the flashlight is pointing toward the nebula and making the gas glow. We think this is part of a filament that may be even more extended than this, but we only see the part of the filament that is illuminated by the beamed emission from the quasar.”


Chemical imaging brings cancer tissue analysis into the digital age

Eliminates delay of weeks for interpretation by histology specialists
Imperial College London researchers have developed a new method for analyzing biological samples based on their chemical makeup that could transform the way medical scientists examine diseased tissue.

When tests are carried out on a patient’s tissue today, such as looking for cancer, the test has to be interpreted by a histology specialist, which can take weeks to get a full result.

Scientists have proposed using mass spectrometry imaging (MSI), which uses technologies that reveal how hundreds or thousands of chemical components are distributed in a tissue sample. But currently proposed MSI workflows are subject to several limitations, including nonoptimized raw data preprocessing, imprecise image coregistration, and limited pattern recognition capabilities.



Physicists create synthetic magnetic monopoles

Nearly 85 years after pioneering theoretical physicist Paul Dirac predicted the possibility of their existence, scientists have created, identified and photographed synthetic magnetic monopoles.
The groundbreaking accomplishment, described by a paper in Nature, paves the way for the detection of the particles in nature, which would be a revolutionary development comparable to the discovery of the electron, according to the scientists.

“The creation of a synthetic magnetic monopole should provide us with unprecedented insight into aspects of the natural magnetic monopole — if indeed it exists,” said Amherst College Physics Professor David S. Hall.

Ordinarily, magnetic poles come in pairs: they have both a north pole and a south pole. As the name suggests, however, a magnetic monopole is a magnetic particle possessing only a single, isolated pole — a north pole without a south pole, or vice versa. In 1931, Dirac published a paper that explored the nature of these monopoles in the context of quantum mechanics. Despite extensive experimental searches since then, no observation of a naturally-occurring magnetic monopole has yet been confirmed and this ground breaking research finally proves another of Dirac's predictions. Of note, it was Dirac who predicted, based on his mathematical model of the electron, the existence of positrons, which was found only few years later to be a real particle, and now widely used including in medical imaging. 

A brain area unique to humans is linked to strategic planning/decision making/multitasking

Oxford University researchers have identified a specific area of the human brain that appears to be unlike anything in the brains of some of our closest relatives.

MRI imaging of 25 adult volunteers was used to identify key components in the area of the human brain called the ventrolateral frontal cortex, and how these components were connected up with other brain areas. The results were then compared with equivalent MRI data from 25 macaque monkeys.
The ventrolateral frontal cortex area of the brain is involved in many of the highest aspects of cognition and language, and is only present in humans and other primates.

Some parts are implicated in psychiatric conditions like ADHD, drug addiction or compulsive behavior disorders. Language is also affected when other parts are damaged after stroke or neurodegenerative disease.

Franz-Xaver Neubert et al. Comparison of human ventral frontal cortex areas for cognitive control and language with areas in monkey frontal cortex, Neuron, 2014

Nanoparticle pinpoints blood-vessel plaques

A step toward identifying plaques vulnerable to rupture that causes heart attack and stroke
A team of researchers led by scientists at Case Western Reserve University has developed a multifunctional nanoparticle that enables magnetic resonance imaging (MRI) to pinpoint blood vessel plaques caused by atherosclerosis. The technology is a step toward creating a non-invasive method of identifying plaques vulnerable to rupture — the cause of heart attack and stroke — in time for treatment.

Currently, doctors can identify only blood vessels that are narrowing due to plaque accumulation. A doctor makes an incision and slips a catheter inside a blood vessel in the arm, groin or neck. The catheter emits a dye that enables X-rays to show the narrowing.

The Case Western Reserve researchers report in the journal Nano Letters that a nanoparticle built from the rod-shaped tobacco mosaic virus, commonly found on tobacco, locates and illuminates plaque in arteries more effectively and with a tiny fraction of the dye.

More importantly, the work shows that the tailored nanoparticles home in on plaque biomarkers. That opens the possibility that particles can be programmed to identify vulnerable plaques from stable ones, something untargeted dyes alone cannot. Steinmetz, a specialist in bioengineering plant viruses, teamed with Xin Yu, a professor of biomedical engineering, who specializes in developing MRI techniques to investigate cardiovascular diseases. They created a device that transports and concentrates imaging agents on plaques.

Elongated nanoparticles have a higher probability of being pushed out of the central blood flow and targeting the vessel wall compared to spheres. The elongated shape also allows more stable attachment to the plaque, the researchers said.

The virus surface is modified to carry short chains of amino acids, called peptides, that make the virus stick where plaques are developing or already exist. Luyt and Simpson synthesized the peptides.
“The binding allows the particle to stay on the site longer, whereas the sheer force is more likely to wash away a sphere, due to its high curvature,” said Yu, an appointee of the Case School of Engineering. 

Mimicking atherosclerosis with blood cells on a microchip

Georgia Institute of Technology scientists have engineered a microchip coated with blood vessel cells. The objective: learn more about the conditions under which nanoparticles accumulate in the plaque-filled arteries of patients with atherosclerosis, the underlying cause of myocardial infarction and stroke.

They coated microchips with a thin layer of endothelial cells, which make up the interior surface of blood vessels.

In healthy blood vessels, endothelial cells act as a barrier to keep foreign objects out of the bloodstream. But at sites prone to atherosclerosis, the endothelial barrier breaks down, allowing things to move in and out of arteries that shouldn’t.


Wearable glasses help surgeons view cancer​​​​​​​​ cells in real time

      Reduce the need for costly additional surgical procedures

Washington University School of Medicine scientists have developed a wearable display to help surgeons visualize cancer cells, which glow blue when viewed through the eyewear.The wearable technology was used during surgery for the first time last week at Alvin J. Siteman Cancer Center at Barnes-Jewish HospitalCancer cells are notoriously difficult to see, even under high-powered magnification. The glasses are designed to make it easier for surgeons to distinguish cancer cells from healthy cells, helping to ensure that no stray tumor cells are left behind during surgery.

  

 Brain signals from a primate directly move paralyzed limbs in another primate ‘avatar’

    Taking brain-machine interfaces (BMI) to the next level, new research may help paralyzed people move their own limb just by thinking about it. Previous research has been limited to controlling external devices, such as robots or synthetic avatar armsIn a paper published online Feb. 18 in Nature Communications, Maryam Shanechi, assistant professor of electrical and computer engineering at Cornell University, working with Ziv Williams, assistant professor of neurosurgery at Harvard Medical School, and colleagues describe a cortical-spinal prosthesis that directs “targeted movement” in paralyzed limbs. The research team developed and tested a prosthesis that connects two subjects (monkeys) by enabling one subject to send its recorded neural activity to control limb movements in a different subject that is temporarily sedated. The demonstration is a step forward in making brain-machine interfaces for paralyzed humans to control their own limbs using their brain activity alone.
  
  The concept: when paralyzed patients imagine or plan a movement, neurons in the brain’s motor cortical areas still activate, even though the communication link between the brain and muscles is broken. By implanting sensors in these brain areas, neural activity could be recorded and translated to the patient’s desired movement using a mathematical transform called the decoder. These interfaces could allow patients to generate movements directly with their thoughts.

      

Imaging small biomolecules inside live cells

      
Researchers at Columbia University have made a significant step toward visualizing small biomolecules inside living biological systems with minimum disturbance, a longstanding goal in the scientific community.

In a study published March 2nd in Nature Methods, Assistant Professor of Chemistry Wei Min’s research team has developed a general method to image a broad spectrum of small biomolecules, such as small molecular drugs and nucleic acids, amino acids, lipids for determining where they are localized and how they function inside cells.

When studying biological functions of a molecule in complex and mysterious cells, researchers typically label the molecules of interest with fluorophores, a kind of molecules that glow when illuminated.

Using a fluorescence microscope, common in research labs, the fluorophore-tagged molecules can be located and tracked with high precision. The invention of green fluorescent protein (GFP), in 1994, compatible with imaging inside live cells and animals, has since made fluorescence microscopy even more popular.

However, when it comes to small biomolecules, fluorophore tagging is problematic, because the fluorophores are almost always larger or comparable in size to the small molecules of interest. As a result, they often disturb the normal functions of these small molecules with crucial biological roles.


First direct evidence of cosmic inflation

Telescope at the South Pole finds twists in microwave-light remnants from the Big Bang, showing evidence for inflation and gravitational waves

Researchers from the BICEP2 telescope collaboration announced Monday the first direct evidence for cosmic inflationThe inflation theory posits that almost 14 billion years ago, the universe we inhabit burst into existence in an extraordinary event that initiated the Big Bang. In the first fleeting fraction of a second, the universe expanded exponentially, stretching far beyond the current view of our best telescopes.

Their data also represent the first images of gravitational waves, or ripples in space-time. These waves have been described as the “first tremors of the Big Bang.” Finally, the data confirm a deep connection between quantum mechanics and general relativity.

“Detecting this signal is one of the most important goals in cosmology today. A lot of work by a lot of people has led up to this point,” said John Kovac (Harvard-Smithsonian Center for Astrophysics), leader of the BICEP2 collaboration.

When asked to comment on the implications of this discovery, Harvard theorist Avi Loeb said, “This work offers new insights into some of our most basic questions: Why do we exist? How did the universe begin? These results are not only a smoking gun for inflation, they also tell us when inflation took place and how powerful the process was.”

BICEP2 is the second stage of a coordinated program, the BICEP and Keck Array experiments, which has a co-principal-investigator (PI) structure. The four PIs are John Kovac (Harvard), Clem Pryke (UMN), Jamie Bock (Caltech/JPL), and Chao-Lin Kuo (Stanford/SLAC). All have worked together on the present result, along with talented teams of students and scientists.
Other major collaborating institutions for BICEP2 include the University of California at San Diego, the University of British Columbia, the National Institute of Standards and Technology (NIST), the University of Toronto, Cardiff University, and Commissariat à l’Energie Atomique.

New live-cell printing technology improves on inkjet printing

A new way to print living cells onto any surface and in almost any shape has been developed by researchers led by Houston Methodist Research Institute nanomedicine faculty member Lidong Qin.
Unlike a similar inkjet printing process, almost all cells survive.

The new process, called Block-Cell-Printing (BloC-Printing), produces 2-D cell arrays in half an hour, prints the cells as close together as 5 microns (most animal cells are 10 to 30 microns wide), and allows the use of many different cell types.

“Cell printing is used in so many different ways now — for drug development and in studies of tissue regeneration, cell function, and cell-cell communication,” Qin said. “Such things can only be done when cells are alive and active. A survival rate of 50 to 80 percent is typical as cells exit the inkjet nozzles.

“By comparison, we are seeing close to 100 percent of cells in BloC-Printing survive the printing process.” BloC-Printing manipulates microfluidic physics to guide living cells into hook-like traps in the silicone mold. Cells flow down a column in the mold, past trapped cells to the next available slot, eventually creating a line of cells in a grid.

The position and spacing of the traps and the shape of the channel navigated by the cells is fully configurable during the mold’s creation. When the mold is lifted away, the living cells remain behind, adhering to the growth medium or other substrate, in prescribed formation.

Wearable ‘neurocam’ records scenes when it detects user interest

Keio University scientists have developed a “neurocam” — a wearable camera system that detects emotions, based on an analysis of the user’s brainwaves. The hardware is a combination of Neurosky’s Mind Wave Mobile and a customized brainwave sensor. The algorithm is based on measures of “interest” and “like” developed by Professor Mitsukura and the neurowear team.

The users interests are quantified on a range of 0 to 100. The camera automatically records five-second clips of scenes when the interest value exceeds 60, with timestamp and location, and can be replayed later and shared socially on Facebook.

The researchers plan to make the device smaller, more comfortable, and fashionable to wear. It is not a stretch to imagine a future dating service making use of this gadget that could literally "read" the wearer's mind!

A new micro-robotic technique for 3D-printing tissues

A new magnetic micro-robotic technique for assembling components of the complex materials used in tissue engineering* and 3D printing of cell materials has been developed by Researchers at Brigham and Women’s Hospital (BWH) and Carnegie Mellon University.
Described in Nature Communications, the technique allows for precise construction of individual cell-encapsulating hydrogels (such as cell blocks).

Described in the Jan. 28, 2014, issue of Nature Communicationsthe research was conducted by Savas Tasoglu, PhD, MS, research fellow in the BWH Division of Renal Medicine, and Utkan Demirci, PhD, MS, associate professor of Medicine in the Division of Biomedical Engineering, part of the BWH Department of Medicine, in collaboration with Eric Diller, PhD, MS, and Metin Sitti, PhD, MS, professor in the Department of Mechanical Engineering, Carnegie Mellon University.

The current process for “bioprinting” cells for tissues or organs (such as a pancreas) is limited because the process can’t be modified  or reversed. For example, misplacement of an ejected droplet or clogging can cause bioprinting to fail, the researchers say in the paper.

“Moreover, simultaneous coding of rigid and soft micro-components into 3D functional materials has presented a significant challenge,” the researchers said in the paper.


New type of MRI ‘whole body’ scan could improve treatment of bone-marrow cancer

A new type of magnetic resonance imaging (MRI) scan could improve care for a type of cancer called myeloma and reduce reliance on bone marrow biopsies, which can be painful for patients and often fail to show doctors how far the disease has spread.

The research, published Feb. 18 in the journal Radiology, was carried out by researchers at The Institute of Cancer Research, London, and The Royal Marsden NHS Foundation Trust. The new whole-body, diffusion-weighted MRI scans showed the spread of cancer throughout the bone marrow of patients with myeloma — one of the most common forms of blood cancer — more accurately than standard tests. The scans also showed whether the patients were responding to cancer treatments.

In the study, 26 patients had whole-body, diffusion-weighted MRI scans before and after treatment. In 86% of cases, experienced doctors trained in imaging were able to correctly identify whether patients responded to treatment. The doctors also correctly identified those patients who weren’t responding to treatment 80% of the time.

Using the scanning technique, doctors could pinpoint exactly where the cancer was in the bones, with the results available immediately. Conventional tests include bone marrow biopsies and blood tests but neither shows accurately where the cancer is present in the bones.
The researchers also assessed the visible changes on the MRI scans, using a measurement called the Apparent Diffusion Coefficient (ADC), which records how restricted water movement is within tissues. Changes in this measurement correctly identified treatment response for 24 of 25 myeloma patients.

The new scan was able to visualize cancer in almost all bones in the body, with only the skull remaining difficult to image, partly because of metal dental implants and fillings. The researchers also found the new methods were suitable for more patients than conventional tests; for example, seven patients had bone marrow biopsies but their samples were found to be inadequate for analysis. Performing another biopsy could be traumatic and painful, and may not provide any new information.

      
      

Magnetic medicine: nanoparticles and magnetic fields train immune cells to fight cancer in mice

Johns Hopkins researchers have trained the immune systems of mice to fight melanoma, a deadly skin cancer, by using nanoparticles designed to target cancer-fighting immune cells,  The experiments, described in ACS Nano February 24, represent a significant step toward using nanoparticles and magnetism to treat a variety of conditions, the researchers say.

“By using small enough particles, we could, for the first time, see a key difference in cancer-fighting cells, and we harnessed that knowledge to enhance the immune attack on cancer,” said Jonathan Schneck, M.D., Ph.D., a professor of pathology, medicine and oncology at the Johns Hopkins University School of Medicine‘s Institute for Cell Engineering.

Schneck’s team has pioneered the development of artificial white blood cells (“artificial antigen-presenting cells” or aAPCs), which show promise in training animals’ immune systems to fight diseases such as cancer. To do that, the aAPCs must interact with immune cells known as naive T cells that are already present in the body, awaiting instructions about which specific invader they will battle.

The aAPCs bind to specialized receptors on the T cells’ surfaces, “presenting” the T cells with distinctive proteins called antigens. This process activates the T cells, programming them to battle a specific threat such as a virus, bacteria, or tumor, as well as to make more T cells.

Magnetic field-based cell clustering activates T cells

To see whether there indeed was a relationship between activation and receptor clustering, Perica applied a magnetic field to the cells, causing the iron-based nano-aAPCs to attract one another and cluster together, bringing the receptors with them. The clustering did indeed activate the naive T cells, and it made the activated cells even more active — effectively ramping up the normal immune response.

To examine how the increased activation would play out in living animals,  Perica tested the impact of these smaller particles.treated a sample of T cells with nano-aAPCs targeting those T cells that were programmed to battle melanoma. The researchers next put the treated cells under a magnetic field and then put them into mice with skin tumors.

The tumors in mice treated with both nano-aAPCs and magnetism stopped growing, and by the end of the experiment, they were about 10 times smaller than those of untreated mice, the researchers found. In addition, they report, six of the eight magnetism-treated mice survived for more than four weeks showing no signs of tumor growth, compared to zero of the untreated mice.

“We were able to fine-tune the strength of the immune response by varying the strength of the magnetic field and how long it was applied, much as different doses of a drug yield different effects,” says Perica. “We think this is the first time magnetic fields have acted like medicine in this way.”
In addition to its potential medical applications, Perica notes that combining nanoparticles and magnetism may give researchers a new window into fundamental biological processes. “In my field, immunology, a major puzzle is how T cells pick out the antigen they’re targeting in a sea of similar antigens in order to find and destroy a specific threat,” he says. “Receptors are key to that action, and the nano-aAPCs let us detect what the receptors are doing.”

“We have a bevy of new questions to work on now: What’s the optimal magnetic ‘dose’? Could we use magnetic fields to activate T cells without taking them out of the body? And could magnets be used to target an immune response to a particular part of the body, such as a tumor’s location?” Schneck adds. “We’re excited to see where this new avenue of research takes us.”

First comprehensive atlas of human gene activity released

A large international consortium of researchers has produced the first comprehensive, detailed map of the way genes work across the major cells and tissues of the human body. The findings describe the complex networks that govern gene activity, and the new information could play a crucial role in identifying the genes involved with disease.

“Now, for the first time, we are able to pinpoint the regions of the genome that can be active in a disease and in normal activity, whether it’s in a brain cell, the skin, in blood stem cells or in hair follicles,” said Winston Hide, associate professor of bioinformatics and computational biology at Harvard School of Public Health (HSPH) and one of the core authors of the main paper in Nature.
“This is a major advance that will greatly increase our ability to understand the causes of disease across the body.”

The research is outlined in a series of papers published March 27, 2014, two in the journal Nature and 16 in other scholarly journals. The work is the result of years of concerted effort among 250 experts from more than 20 countries as part of FANTOM 5 (Functional Annotation of the Mammalian Genome). The FANTOM project, led by the Japanese institution RIKEN, is aimed at building a complete library of human genes.

Alistair R. R. Forrest et al., A promoter-level mammalian expression atlas, Nature, 2014, DOI: 10.1038/nature13182

MIT engineers design hybrid living/nonliving materials

MIT engineers have coaxed bacterial cells to produce biofilms that can incorporate nonliving materials, such as gold nanoparticles and quantum dots.

These “living materials” combine the advantages of live cells — which respond to their environment, produce complex biological molecules, and span multiple length scales — with the benefits of nonliving materials, which add functions such as conducting electricity or emitting light.

This approach could one day be used to design more complex devices such as solar cells, self-healing materials, or diagnostic sensors, says Timothy Lu, an MIT assistant professor of electrical engineering andbiological engineering. Lu is the senior author of a paper describing this innovation in the March 23 issue of Nature MaterialsThe researchers also demonstrated that the cells can even coordinate with each other to control the composition of the biofilm.

These hybrid materials could be worth exploring for use in energy applications such as batteries and solar cells, Lu says. The researchers are also interested in coating the biofilms with enzymes that catalyze the breakdown of cellulose, which could be useful for converting agricultural waste to biofuels. Other potential applications include diagnostic devices and scaffolds for tissue engineering.

     

 Electric ‘thinking cap’ can help you learn faster, better

In a new study published in the Journal of NeuroscienceVanderbilt psychologists  show that it is possible to learn through the application of a mild electrical current to the brain, and that this effect can be enhanced or depressed depending on the direction of the current.

The medial-frontal cortex is believed to be the part of the brain responsible for the instinctive “Oops!” response we have when we make a mistake. Previous studies have shown that a voltage spike originates from this area of the brain milliseconds after a person makes a mistake. Robert Reinhart, a Ph.D. candidate, and Geoffrey Woodman, assistant professor of psychology, wanted to test the idea that this activity influences learning because it allows the brain to learn from our mistakes.

Reinhart and Woodman set out to test several hypotheses, including:
1) It is possible to control the brain’s electrophysiological response to mistakes.
2) Its effect could be intentionally regulated up or down depending on the direction of an electrical current applied to it. 

After 20 minutes of stimulation, subjects were given a learning task that involved figuring out by trial and error which buttons on a game controller corresponded to specific colors displayed on a monitor. The task was made more complicated by occasionally displaying a signal for the subject not to respond—sort of like a reverse “Simon Says.” For even more difficulty, they had less than a second to respond correctly, providing many opportunities to make errors—and, therefore, many opportunities for the medial-frontal cortex to fire.

The researchers measured the EEG (electrical brain activity) of each participant. This allowed them to watch as the brain changed at the very moment participants were making mistakes, and most importantly, allowed them to determine how these brain activities changed under the influence of electrical stimulation.

Stimulating or inhibiting learning

When anodal current was applied, the EEG brain-activity spike was almost twice as large on average and was significantly higher in a majority of the individuals tested (about 75 percent of all subjects across four experiments). This was reflected in their behavior; they made fewer errors and learned from their mistakes more quickly than they did after the sham stimulus.

When cathodal current was applied, the researchers observed the opposite result: The spike was significantly smaller, and the subjects made more errors and took longer to learn the task. “So when we up-regulate that process, we can make you more cautious, less error-prone, more adaptable to new or changing situations—which is pretty extraordinary,” Reinhart said.

The effect was not noticeable to the subjects — their error rates only varied about 4 percent either way, and the behavioral adjustments adjusted by a matter of only 20 milliseconds — but they were plain to see on the EEG. “This success rate is far better than that observed in studies of pharmaceuticals or other types of psychological therapy,” said Woodman. The researchers found that the effects of a 20-minute stimulation did transfer to other tasks and lasted about five hours. The implications of the findings extend beyond the potential to improve learning. The process may also have clinical benefits in the treatment of conditions like schizophrenia and ADHD, which are associated with performance-monitoring deficits, the researchers suggest.

R. M. G. Reinhart, G. F. Woodman, Causal Control of Medial-Frontal Cortex Governs Electrophysiological and Behavioral Indices of Performance Monitoring and Learning, Journal of Neuroscience, 2014, DOI: 10.1523/JNEUROSCI.5421-13.2014

Magnetically controlled nanoparticles cause cancer cells to self-destruct

Researchers at Lund University in Sweden have developed a technique to use magnetically controlled nanoparticles to force tumor cells to “self-destruct.” without harming surrounding tissue, as with radiotherapy, and tissues elsewhere in the body, as with chemotherapy. “Our technique is able to attack only the tumor cells,” said Enming Zhang, first author of the study.

Inducing cell suicide

The technique involves getting the nanoparticles into a tumor cell, where they bind to lysosomes, which can break down foreign substances that have entered a cell. They can also break down the entire cell through a process known as apoptosis (controlled cell death), a type of destruction where damaged cells dissolve themselves.

The researchers used superparamagnetic nanoparticles of iron oxide. Once the particles are inside the cancer cells, the cells are exposed to an external magnetic field, and the nanoparticles begin to rotate in a way that causes the lysosomes to start destroying the cells.

Previous attempts to use superparamagnetic nanoparticles.have focused on using the external field to create heat that kills the cancer cells. The problem with this is that the heat can cause inflammation that risks harming surrounding, healthy tissue. The new method, on the other hand, in which the rotation of the magnetic nanoparticles can be controlled, only affects the tumor cells that the nanoparticles have entered.

The new technique is primarily intended for cancer treatment, but according to the researchers, it can be used for other diseases, including autoimmune diseases such as type 1 diabetes, in which the immune system attacks the body’s own insulin production.

The researchers at Lund University have a patent pending for their technique with the rotating nanoparticles. However, a lot of work remains before it can be transferred from the laboratory to clinical trials on patients.

The study, a collaboration between physicists, chemists, engineers and doctors from Sweden, Germany and the U.S., was published in the journal ACS Nano.

A blueprint for how to build a human brain

Researchers at the Allen Institute for Brain Science have generated a blueprint for how to build a human brain at unprecedented anatomical resolution.

This first major report using data from the  the BrainSpan Atlas of the Developing Human Brain is published in the journal Nature this week. The data provide insight into diseases like autism that are linked to early brain development, and to the origins of human uniqueness. The rich data set is publicly available via the Allen Brain Atlas data portal.

“Knowing where a gene is expressed in the brain can provide powerful clues about what its role is,” says Ed Lein, Investigator at the Allen Institute for Brain Science.  “This atlas gives a comprehensive view of which genes are on and off in which specific nuclei and cell types while the brain is developing during pregnancy. This means that we have a blueprint for human development: an understanding of the crucial pieces necessary for the brain to form in a normal, healthy way, and a powerful way to investigate what goes wrong in disease.”

This paper represents the first major report to make use of data collected for the BrainSpan Atlas of the Developing Human Brain, a science consortium initiative that seeks to create a map of the transcriptome across the entire course of human development.

“This atlas is already transforming the way scientists approach human brain development and neurodevelopmental disorders like autism and schizophrenia,” said Thomas R. Insel, Director of the National Institute of Mental Health.”

The researchers pointed to autism as a disorder with particularly pertinent links to early brain development. The research team used the BrainSpan Atlas to examine a number of genes linked to autism in prior scientific studies during development.

“We used the maps we created to find a hub of genetic action that could be linked to autism—and we found one,” says Lein. “These genes were associated with the newly generated excitatory neurons in the cortex, the area of the brain that is responsible for many of the cognitive features affected in autism such as social behavior. This discovery is an exciting example of the ability of the BrainSpan Atlas to generate meaningful hypotheses about the origins of brain developmental disorders.”

What makes humans unique?

Understanding what makes humans unique involves deciphering a complex puzzle—one that begins during the earliest phases of development. The richness of the BrainSpan Atlas gives scientists a new set of tools to assess how the human brain develops compared to other species.

“We know that some important regions of the genome show striking sequence differences in humans compared to other species,” says Lein. “Since where a gene is expressed in the brain can give insight into its function, we can use our map to begin to figure out the roles of those genes in making humans distinct. Our analysis of the data showed that these genes are enriched in the frontal cortex, as well as in several specific specialized cell types including inhibitory GABAergic interneurons and neurons of the transient subplate zone that serves as a scaffold during early circuit formation.  These features are all known to be expanded or show developmental differences in humans compared to other species, so our data gives unprecedented clues about the molecular underpinnings of what makes human neocortex unique.”

The BrainSpan Atlas enables researchers around the world to conduct research and ask questions about the early human brain that many would not be able to do otherwise, due to the highly limited availability of prenatal tissues.

The founding principal investigators in the consortium behind the BrainSpan project include Ed Lein and Michael Hawrylycz at the Allen Institute for Brain Science, Nenad Sestan and Mark Gerstein at Yale University, Jim Knowles at USC , Pat Levittat at The Saban Research Institute of Children’s Hospital Los Angeles and USC, Dan Geschwind at UCLA, and Bruce Fischl at Massachusetts General Hospital.

Einstein’s skepticism about quantum mechanics may lead to ultra-secure Internet


Einstein’s skepticism* about quantum mechanics may lead to an ultra-secure Internet, suggests a new paper by researchers from Swinburne University of Technology and Peking University. Associate Professor Margaret Reid from Swinburne’s Center for Quantum and Optical Science said Einstein’s reservations about quantum mechanics were highlighted in a phenomenon known as “spooky action at a distance,” which is the strange way entangled particles stay connected even when separated by large distances.

“Until now the real application of this has been for messages being shared between two people securely without interception, regardless of the spatial separation between them,” Reid said.

“In this paper, we give theoretical proof that such messages can be shared between more than two people and may provide unprecedented security for a future quantum internet.”

In the 1990s, scientists realized you can securely transmit a message through encrypting and using a shared key generated by entanglement to decode the message from the sender and receiver. Using the quantum key meant the message was completely secure from interception during transmission.

Sending entanglement to a larger number of people means the key can be distributed among all the receiving parties, so they must collaborate to decipher the message, which Reid said makes the message even more secure. “We found that a secure message can be shared by up to three to four people, opening the possibility to the theory being applicable to secure messages being sent from many to many. “The message will also remain secure if the devices receiving the message have been tampered with, because of the nature of entanglement.”

“This proof is at the fundamental theoretical level only and has not yet been experimentally observed,” Reid explained to KurzweilAI. While commercial application is some time away, it “provides the potential for a strong form of multi-party quantum cryptography, where there is security against infiltrators tampering with devices at the receiving stations.”


When scientists transplanted human neural stem cells into mice with multiple sclerosis (MS), within a remarkably short period of time, 10 to 14 days, the mice had regained motor skills. Six months later, they showed no signs of slowing down. Results from the study demonstrate that the mice experience at least a partial reversal of symptoms. Immune attacks are blunted, and the damaged myelin is repaired, explaining their dramatic recovery. The finding, which uncovers potential new avenues for treating MS, was published May 15, 2014 in the journalStem Cell Reports (open access)  Ronald Coleman (a graduate student of Jeanne Loring, Ph.D., co-senior author and director of the Center for Regenerative Medicine at The Scripps Research Institute and co-first author on the publication) changed the normal protocol and grew the neural stem cells so they were less crowded on a Petri dish than usual. That yielded a human neural stem cell type that turned out to be extremely potent. The experiments have since been successfully repeated with cells produced under the same conditions, but by different laboratories.


Brain-controlled airplanes


Pilots of the future could fly a plane by just thinking commands, say scientists at the Institute for Flight System Dynamics atTechnische Universität München (TUM) and Technische Universität Berlin (TU Berlin) involved in the EU-fundedBrainflight project.

The system uses electroencephalography (EEG) to detect brain waves. An algorithm developed by scientists from Team PhyPA(Physiological Parameters for Adaptation) at TU Berlin deciphers electrical potentials and converts them into control commands.

“A long-term vision of the project is to make flying accessible to more people,” explains aerospace engineer Tim Fricke, who heads the project at TUM. “With brain control, flying could become easier.
This would reduce the work load of pilots and thereby increase safety. In addition, pilots would have more freedom of movement to manage other manual tasks in the cockpit.”


Milky Way may have 100 million life-giving planets


“It seems highly unlikely that we are alone.”
There are some 100 million other places in the Milky Way galaxy that could support life above the microbial level, reports a group of astronomers in the journal Challenges (open access), based on a new computation method to examine data from planets orbiting other stars in the universe.

“This study does not indicate that complex life exists on that many planets; we’re saying that there are planetary conditions that could support it, according to the paper’s authors*.  “Complex life doesn’t mean intelligent life — though it doesn’t rule it out or even animal life — but simply that organisms larger and more complex than microbes could exist in a number of different forms,” the researchers explain.

The scientists surveyed more than 1,000 planets and used a formula that considers planet density, temperature, substrate (liquid, solid or gas), chemistry, distance from its central star and age. From this information, they developed and computed the Biological Complexity Index (BCI).
The BCI calculation revealed that 1 to 2 percent of the planets showed a BCI rating higher than Europa, a moon of Jupiter thought to have a subsurface global ocean that may harbor forms of life. With about 10 billion stars in the Milky Way galaxy, the BCI yields 100 million plausible planets.
The authors cite one study that suggests that “some exoplanets may be more optimally suited for life than Earth. … Such ‘superhabitable’ worlds would likely be larger, warmer, and older, orbiting dwarf stars.”

“It seems highly unlikely that we are alone,” say the researchers. “We are likely so far away from life at our level of complexity that a meeting with such alien forms might be improbable for the foreseeable future.”


Researchers create miniature human retina in a dish

Johns Hopkins researchers have created a miniature human retina in a dish that can sense light. The work, reported online June 10 in the journal Nature Communications, “advances opportunities for vision-saving research and may ultimately lead to technologies that restore vision in people with retinal diseases,” says study leader M. Valeria Canto-Soler, Ph.D., an assistant professor of ophthalmology at the Johns Hopkins University School of Medicine.

The achievement emerged from experiments with human induced pluripotent stem cells (iPS). While the system doesn’t yet produce images, it could eventually enable genetically engineered retinal cell transplants that halt or even reverse a patient’s march toward blindness, the researchers say.

The iPS cells are adult cells that have been genetically reprogrammed to their most primitive state. Under the right circumstances, they can develop into most or all of the 200 cell types in the human body. In this case, the Johns Hopkins team turned them into retinal progenitor cells destined to form light-sensitive retinal tissue that lines the back of the eye.

Canto-Soler says that the newly developed system gives them the ability to generate hundreds of mini-retinas at a time directly from a person affected by a particular retinal disease such as retinitis pigmentosa. This provides a unique biological system to study the cause of retinal diseases directly in human tissue, instead of relying on animal models.


Engineered red blood cells could carry therapeutic or diagnostic payloads


Whitehead Institute scientists and associates have modified red blood cells (RBCs) to carry a range of valuable therapeutic and diagnostic payloads — such as drugs, vaccines, and disease-detecting imaging agents  — for delivery to specific sites throughout the body.

“We wanted to create high-value red cells that do more than simply carry oxygen,” says Whitehead Founding Member Harvey Lodish, who collaborated with Whitehead Member Hidde Ploegh in this pursuit. So they modified the genes and enzymes in mouse and human RBCs in culture (in the lab). The work, published this week in the Proceedings of the National Academy of Sciences (PNAS), combines Lodish’s expertise in the biology of red blood cells (RBCs) with biochemical methods developed in Ploegh’s lab.

RBCs are an attractive vehicle for potential therapeutic applications, the researchers say. They are more numerous than any other cell type in the body. They have a long lifespan (up to 120 days in circulation). And during RBC production, the progenitor cells that eventually mature to become RBCs jettison their nuclei and all DNA. Without a nucleus, a mature RBC lacks any genetic material or any signs of earlier genetic manipulation that could result in tumor formation or other adverse effects.

Wide range of medical uses

The researchers suggest that the applications are potentially vast, including:
·         Bind and remove bad cholesterol from the bloodstream.
·         Carry clot-busting proteins to treat ischemic strokes or deep-vein thrombosis.
·         Deliver anti-inflammatory antibodies to alleviate chronic inflammation.
·        Suppress the unwanted immune response that often accompanies treatment with protein-based therapies.
·         Neutralize a toxin. “Because the modified human red blood cells can circulate in the body for up to four months, one could envision a scenario in which the cells are used to introduce antibodies that neutralize a toxin,” says Ploegh. “The result would be long-lasting reserves of antitoxin antibodies.” That’s why the U.S. military and its Defense Advanced Research Projects Agency (DARPA) is supporting the research at Whitehead in the interest of developing treatments or vaccines effective against biological weapons.


Google Glass app aims to improve surgeon training in Stanford University Medical School

CrowdOptic is working with the Department of Cardiothoracic Surgery at Stanford University Medical Center to use CrowdOptic’s Google Glass software to help improve resident training in complex surgical procedures, the company has announced.

CrowdOptic’s app gives a Google Glass wearer — such as a surgeon — access to what another user — such as a resident performing an operation — is seeing, simply by looking in the resident’s direction, in this case.

Traditionally, the restricted view in the operating room has made it next to impossible for an attending surgeon to see the exact field of view of a trainee, complicating the process of providing essential feedback on techniques.

CrowdOptic CEO Jon Fisher said he hopes that this technology will “offer a paradigm shift in surgical training, especially in the highly complex area of cardiothoracic training, where a major challenge is creating an environment in which an attending surgeon can provide direct visual feedback to residents conducting operations.” CrowdOptic will be deployed in Stanford Medical Center, where faculty and student teams evaluate training in a variety of surgical settings.

CrowdOptic’s context-aware apps engage fans and live audiences, producing crowd sourced content for live broadcasts. CrowdOptic analyzes the best views and footage, obtained through devices such as Google Glass, then streams the live feeds directly to arena video boards, and even allows fans to see each other’s views.

Implanted neuronal stem cells generate neurons and synapses, becoming a functioning part of mouse brain

Scientists at the Luxembourg Centre for Systems Biomedicine (LCSB) of the University of Luxembourg have grafted induced neuronal stem cells (iNSC) into the brains of mice, with long-term functionality and stability, for the first time. Six months after implantation, the new neurons, reprogrammed from skin cells, became fully and functionally integrated into the brain, creating synapses and glial cells.

This successful implantation of neurons raises hope for future therapies for neurodegenerative diseases, replacing sick neurons with healthy ones — in the brains of Parkinson’s disease patients, for example. However, “successes in human therapy are still a long way off,” cautioned principal investigator Prof. Jens Schwamborn.

The researchers published their results in Stem Cell Reports (open access).


Fully integrated into the brain, connected by synapses


The treated mice showed no adverse side effects, even six months after implantation into the hippocampus and cortex regions of the brain, according to the researchers. The neurons exhibited normal activity and were fully integrated into the complex network of the brain, connected to the original brain cells via newly formed synapses.
In addition, the iNSCs are not predisposed to tumor formation, as in the case of induced pluripotent stem cells (iPSCs).

“Building upon the current insights, we will now be looking specifically at the type of neurons that die off in the brain of Parkinson’s patients — namely, the dopamine-producing neurons,” Schwamborn said. He said that in the future, implanted neurons could produce dopamine (which is lacking in Parkinson’s) directly in the patient’s brain and transport it to the appropriate sites — constituting an actual cure.

Do gut bacteria control your mind?

Bacteria within you — which outnumber your own cells about 100 times — may be affecting both your cravings and moods to get you to eat what they want, and may be driving you toward obesity. That’s the conclusion of an article published this week in the journal BioEssays by researchers from UC San Francisco, Arizona State University and University of New Mexico from a review of the recent scientific literature.

How your gut microbiome may control you

The diverse community of microbes, collectively known as the gut microbiome, influence human eating behavior and dietary choices to favor consumption of the particular nutrients they grow best on, rather than simply passively living off whatever nutrients we choose to send their way. Some bacterial species prefer fat, and others sugar, for instance. They vie with each other for food and to retain a niche within their ecosystem — your digestive tract — and they also often have different aims than you do when it comes to your own actions. Bacteria may influence your decisions by releasing signaling molecules into your gut. Because the gut is linked to the immune system, the endocrine system, and the nervous system, those signals could influence your physiologic and behavioral responses — and health. Bacteria may be acting through the vagus nerve, which connects 100 million nerve cells from the digestive tract to the base of the brain, changing taste receptors, producing toxins to make you feel bad, and releasing chemical rewards to make you feel good. Certain strains of bacteria increase anxious behavior (in mice). Some strains of bacteria cause stomach cancer and perhaps other cancers. 


First direct brain-to-brain communication between human subjects

EEG and TMS signals enable first successful brain-to-brain transmission
An international team of neuroscientists and robotics engineers have demonstrated the first direct remote brain-to-brain communication between two humans located 5,000 miles away from each other and communicating via the Internet, as reported in a paper recently published in PLOS ONE (open access).

In India, researchers encoded two words (“hola” and “ciao”) as binary strings and presented them as a series of cues on a computer monitor. They recorded the subject’s EEG signals as the subject was instructed to think about moving his feet (binary 0) or hands (binary 1). They then sent the recorded series of binary values in an email message to researchers in France, 5,000 miles away.
There, the binary strings were converted into a series of transcranial magnetic stimulation (TMS) pulses applied to a hotspot location in the right visual occipital cortex that either produced a phosphene (perceived flash of light) or not.

“We wanted to find out if one could communicate directly between two people by reading out the brain activity from one person and injecting brain activity into the second person, and do so across great physical distances by leveraging existing communication pathways,” explains coauthor Alvaro Pascual-Leone, MD, PhD, Director of the Berenson-Allen Center for Noninvasive Brain Stimulation at Beth Israel Deaconess Medical Center (BIDMC) and Professor of Neurology at Harvard Medical School.

A team of researchers from Starlab Barcelona, Spain and Axilum Robotics, Strasbourg, France conducted the experiment. A second similar experiment was conducted between individuals in Spain and France.

“We believe these experiments represent an important first step in exploring the feasibility of complementing or bypassing traditional language-based or other motor/PNS mediated means in interpersonal communication,” the researchers say in the paper. As is usual for scientists, this is an understatement for the future potentials of this technology, where people one day could communicate across the globe using their brain alone, and across various cultural and linguistic barriers.

Slowing down the aging process by ‘remote control’


UCLA biologists have identified a gene that can slow the aging process throughout the entire body when activated remotely in key organ systems.

Working with fruit flies, the life scientists activated a gene called AMPK that is a key energy sensor in cells; it gets activated when cellular energy levels are low.

Increasing the amount of AMPK in fruit flies’ intestines increased their lifespans by about 30 percent — to roughly eight weeks from the typical six — and the flies stayed healthier longer as well. The research, published Sept. 4 in the open-source journal Cell Reports, could have important implications for delaying aging and disease in humans, said David Walker, an associate professor of integrative biology and physiology at UCLA and senior author of the research.

“We have shown that when we activate the [AMPK] gene in the intestine or the nervous system, we see the aging process is slowed beyond the organ system in which the gene is activated,” Walker said.
Walker said that the findings are important because extending the healthy life of humans would presumably require protecting many of the body’s organ systems from the ravages of aging — but delivering anti-aging treatments to the brain or other key organs could prove technically difficult. The study suggests that activating AMPK in a more accessible organ such as the intestine, for example, could ultimately slow the aging process throughout the entire body, including the brain.

Humans have AMPK, but it is usually not activated at a high level, Walker said. “Instead of studying the diseases of aging — Parkinson’s disease, Alzheimer’s disease, cancer, stroke, cardiovascular disease, diabetes — one by one, we believe it may be possible to intervene in the aging process and delay the onset of many of these diseases,” said Walker, a member of UCLA’s Molecular Biology Institute. “We are not there yet, and it could, of course, take many years, but that is our goal and we think it is realistic.

“The ultimate aim of our research is to promote healthy aging in people.”

The fruit fly, Drosophila melanogaster, is a good model for studying aging in humans because scientists have identified all of the fruit fly’s genes and know how to switch individual genes on and off. The biologists studied approximately 100,000 of them over the course of the study.

Lead author Matthew Ulgherait, who conducted the research in Walker’s laboratory as a doctoral student, focused on a cellular process called autophagy, which enables cells to degrade and discard old, damaged cellular components. By getting rid of that “cellular garbage” before it damages cells, autophagy protects against aging, and AMPK has been shown previously to activate this process. Ulgherait studied whether activating AMPK in the flies led to autophagy occurring at a greater rate than usual.

“A really interesting finding was when Matt activated AMPK in the nervous system, he saw evidence of increased levels of autophagy in not only the brain, but also in the intestine,” said Walker, a faculty member in the UCLA College. “And vice versa: Activating AMPK in the intestine produced increased levels of autophagy in the brain — and perhaps elsewhere, too.” Many neurodegenerative diseases, including both Alzheimer’s and Parkinson’s, are associated with the accumulation of protein aggregates, a type of cellular garbage, in the brain, Walker noted.

“Matt moved beyond correlation and established causality,” he said. “He showed that the activation of autophagy was both necessary to see the anti-aging effects and sufficient; that he could bypass AMPK and directly target autophagy.”

Walker said that AMPK is thought to be a key target of metformin, a drug used to treat Type 2 diabetes, and that metformin activates AMPK.

Milestone reached in building replacement kidneys in the lab

Regenerative medicine researchers at Wake Forest Baptist Medical Center in North Carolina have developed what they say is the most successful method to date to keep blood vessels in new human-sized pig kidney organs open and flowing with blood — a major challenge in the quest to build replacement kidneys in the lab. The work is reported in the journal Technology.
“Until now, lab-built kidneys have been rodent-sized and have functioned for only one or two hours after transplantation because blood clots developed,” said Anthony Atala, M.D., director and professor at the Wake Forest Institute for Regenerative Medicine and a senior author on the study.
“In our proof-of-concept study, the [blood] vessels in a human-sized pig kidney remained open during a four-hour testing period. We are now conducting a longer-term study to determine how long flow can be maintained.”

If proven successful, the new method to more effectively coat the blood vessels with endothelial cells (which line the interior surface of blood vessels) could potentially be applied to other complex organs that scientists are working to engineer, including the liver and pancreas, the researchers say.

How to create a replacement kidney that survives

The research is part of a long-term project to use pig kidneys to make scaffolds (support structures)  that could potentially be used to build replacement kidneys for human patients who have end-stage renal disease. The main problem is dealing with blood clotting.
The current process fails within a few hours:
1. To avoid immune rejection, scientists currently first remove all animal cells (proteins and DNA) from the organ —  leaving only the organ structure or “skeleton”(this is known as “decellularization.” A patient’s own cells are then be placed in the scaffold (“recellularization”), making an organ that the patient (theoretically) would not reject.
2. The cell removal process leaves behind an intact network of blood vessels that can potentially supply the new organ with oxygen. However, scientists working to repopulate kidney scaffolds with cells have had problems coating the vessels, so severe clotting has generally occurred within a few hours after transplantation.

The new Wake Forest Baptist approach looks more promising:


How to ‘switch off’ autoimmune diseases

University of Bristol researchers have discovered how to stop cells from attacking healthy body tissue in debilitating autoimmune diseases (such as multiple sclerosis), where the body’s immune system destroys its own tissue by mistake.
The cells were converted from being aggressive to actually protecting against disease.
The study, funded by the Wellcome Trust, was published September 3 in Nature Communications (open access).

The researchers hope the finding will lead to widespread use of “antigen-specific immunotherapy” as a treatment for many autoimmune disorders, including multiple sclerosis (MS), type 1 diabetes, Graves’ disease, and systemic lupus erythematosus (SLE). (An antigen is a substance that generates an adaptive immune response.)

From aggressor to protector

Scientists were able to selectively target the cells that cause autoimmune disease by reducing their aggression against the body’s own tissues and converting them into cells capable of protecting against disease. This type of conversion has been previously applied to allergies (“allergic desensitization”). They also found that effective treatment is achieved by gradually increasing the dose of antigenic fragment injected.

To find out how this type of immunotherapy works, the scientists delved inside the immune cells themselves to see which genes and proteins were turned on or off by the treatment. They found changes in gene expression that help explain how effective treatment leads to conversion of aggressor into protector cells.

By specifically targeting the specific aggressor cells, this immunotherapeutic approach avoids the need for the immune suppressive drugs associated with infections, development of tumors, and disruption of natural regulatory mechanisms.

“Insight into the molecular basis of antigen-specific immunotherapy opens up exciting new opportunities to enhance the selectivity of the approach while providing valuable markers with which to measure effective treatment,” said Professor David Wraith, who led the research. “These findings have important implications for the many patients suffering from autoimmune conditions that are currently difficult to treat.”

MS affects around 2.5 million people worldwide.

First genetic-based tool to detect circulating cancer cells in blood — lights up cancer cells

Northwestern University scientists have demonstrated a simple but powerful tool called NanoFlare that can detect live cancer cells in the bloodstream, potentially long before settling somewhere in the body and forming a dangerous tumor.The NanoFlare technology is the first genetic-based approach that is able to detect live circulating tumor cells out of the complex matrix that is human blood — no easy feat. The NanoFlares are tiny spherical nucleic acids with gold nanoparticle cores outfitted with single-stranded DNA “flares” (glowing markers).In a breast cancer study, the NanoFlares easily entered cells and lit up the cell if a biomarker target was present, even if only a trace amount.“This technology has the potential to profoundly change the way breast cancer in particular and cancers in general are both studied and treated,” said Chad A. Mirkin, PhD, a noted nanomedicine expert and a corresponding author of the study.


Mirkin’s colleagues C. Shad Thaxton, M.D. and Dr. Chonghui Cheng, M.D., both of Northwestern University Feinberg School of Medicine, are also corresponding authors.The research team, in a paper published the week of Nov. 17 in the Proceedings of the National Academy of Sciences(PNAS), reports two key innovations:·         The ability to track tumor cells in the bloodstream based on genetic content located within the cell itself, as opposed to using proteins located on the cell’s surface (current technology). The ability to collect the cells in live form, so they may be studied and used to inform researchers and clinicians as to how to treat a disease — an important step toward personalized medicine


“Cancers are very genetically diverse, and it’s important to know what cancer subtype a patient has,” Mirkin said. “Now you can think about collecting a patient’s cells and studying how those cells respond to different therapies. The way a patient responds to treatment depends on the genetic makeup of the cancer.”Mirkin is the George B. Rathmann Professor of Chemistry in the Weinberg College of Arts and Sciences and professor of medicine, chemical and biological engineering, biomedical engineering and materials science and engineering. 

How it works: A NanoFlare is designed to recognize a specific genetic code snippet associated with a cancer. The core nanoparticle, only 13 nanometers in diameter, enters cells, and the NanoFlare seeks its target. If the genetic target is present in the cell, the NanoFlare binds to it and the reporter “flare” is released to produce a fluorescent signal. The researchers then can isolate those cells.“The NanoFlare turns on a light in the cancer cells you are looking for,” said Thaxton, an assistant professor of urology at Feinberg. “That the NanoFlares are effective in the complex matrix of human blood is a great technical advance. We can find small numbers of cancer cells in blood, which really is like searching for a needle in a haystack.”Once they identified the cancer cells, the researchers were able to separate them from normal cells. This ability to isolate, culture and grow the cancer cells will allow researchers to zero in on the cancer cells that matter to the health of the patient. Most circulating tumor cells may not metastasize, and analysis of the cancer cells could identify those that will.

“This could lead to personalized therapy where we can look at how an individual’s cells respond to different therapeutic cocktails,” said Mirkin, whose lab developed NanoFlares.

Could ibuprofen be an anti-aging medicine?

Ibuprofen, a common over-the-counter drug used to relieve pain and fever, could hold the keys to a longer healthier life, according to a study by researchers at the Buck Institute for Research on Aging.  Publishing in PLoS Genetics (open access) December 18, scientists showed that regular doses of ibuprofen extended the lifespan of yeast, worms and fruit flies.

Brian Kennedy, PhD, CEO of the Buck Institute, said treatments, given at doses comparable to those used in humans, extended lifespan an average of 15 percent in the model organisms. “Not only did all the species live longer, but the treated flies and worms appeared more healthy,” he said.

“The research shows that ibuprofen impacts a process not yet implicated in aging, giving us a new way to study and understand the aging process.” But most importantly, Kennedy said the study opens the door for a new exploration of “anti-aging medicines.”

The work was the result of a collaboration between the Buck Institute and Texas A & M’s  Agrilife program.  Michael Polymenis, PhD, an AgriLife Research biochemist started the work in baker’s yeast and then moved it into worms and flies. Polymenis, who also is a professor in the biochemistry and biophysics department at Texas A&M University, said the three-year project showed that ibuprofen interferes with the ability of yeast cells to pick up tryptophan, an amino acid found in every cell of every organism.

Tryptophan is essential for humans, who get it from protein sources in the diet. “We are not sure why this works, but it’s worth exploring further. This study was a proof of principle, to show that common, relatively safe drugs in humans can extend the lifespan of very diverse organisms,” he said. “Therefore, it should be possible to find others like ibuprofen with even better ability to extend lifespan, with the aim of adding healthy years of life in people.”

Equivalent to a dozen or so human years

Chong He, PhD, a postdoctoral fellow at the Buck Institute and lead author on the paper, said the extended lifespan in the model organisms would be the equivalent to another dozen or so years of healthy living in humans.  “Our preliminary data in the worms showed that ibuprofen also extended their healthspan,” she said. “Healthy worms tend to thrash a lot and the treated worms thrashed much longer than would be normally expected. As they aged, they also swallowed food much faster than expected.”

In C. elegans worms, ibuprofen has also been shown to suppress a phenotype associated with aging, inhibiting the deposition of amyloid β peptide, a marker for Alzheimer disease, the paper notes. Ibuprofen is in the class of compounds known as NSAIDs — nonsteroidal anti-inflammatory drugs used for relieving pain, helping with fever and reducing inflammation.  The World Health Organization includes ibuprofen on their “List of Essential Medications” needed in a basic health system. Although deemed relatively safe and commonly used, ibuprofen can have adverse side effects, particularly in the gastrointestinal tract and the liver at high doses.

      

 Deep neural network rivals primate brain in object recognition

A new study from MIT neuroscientists has found that for the first time, one of the latest generation of “deep neural networks” matches the ability of the primate brain to recognize objects during a brief glance.

Because these neural networks were designed based on neuroscientists’ current understanding of how the brain performs object recognition, the success of the latest networks suggests that neuroscientists have a fairly accurate grasp of how object recognition works, says James DiCarlo, a professor of neuroscience and head of MIT’s Department of Brain and Cognitive Sciences and the senior author of a paper describing the study in the Dec. 18 issue of the open-access journal PLoS Computational Biology.

Primates visually recognize and determine the category of an object even at a brief glance, and to date, this behavior has been unmatched by artificial systems.

Lead author Charles Cadieu and colleagues from MIT measured the brain’s object recognition ability by implanting arrays of electrodes in the inferior temporal cortex of macaque monkeys and in area V4, a part of the visual system that feeds into the that area of the cortex. This allowed the researchers to see the neural representation — the population of neurons that respond — for every object that the animals looked at.

When comparing these results with representations created by the deep neural networks, the accuracy of the model was determined by whether it grouped similar objects into similar clusters within the representation.

This improved understanding of how the primate brain works could lead to better artificial intelligence and provide insight into understanding primate visual processing.
“The fact that the models predict the neural responses and the distances of objects in neural population space shows that these models encapsulate our current best understanding as to what is going on in this previously mysterious portion of the brain,” say the authors.

More processing power and data

Two major factors account for the recent success of this type of neural network, Cadieu says. One is a significant leap in the availability of computational processing power, using relatively inexpensive graphical processing units (GPUs). The second factor is that researchers now have access to large datasets to feed the algorithms to “train” them. These datasets contain millions of images, and each one is annotated by humans with different levels of identification. For example, a photo of a dog would be labeled as animal, canine, domesticated dog, and the breed of dog.

Cadieu says that researchers don’t know much about what exactly allows these networks to distinguish different objects. “That’s a pro and a con,” he says. “It’s very good in that we don’t have to really know what the things are that distinguish those objects. But the big con is that it’s very hard to inspect those networks, to look inside and see what they really did. Now that people can see that these things are working well, they’ll work more to understand what’s happening inside of them.”

DiCarlo’s lab now plans to try to generate models that can mimic other aspects of visual processing, including tracking motion and recognizing three-dimensional forms. They also hope to create models that include the feedback projections seen in the human visual system. Current networks only model the “feedforward” projections from the retina to the IT cortex, but there are 10 times as many connections that go from IT cortex back to the rest of the system.


CAR T-Cell Therapy: Engineering Patients’ Immune Cells to Treat Their Cancers


For years, the cornerstones of cancer treatment have been surgery, chemotherapy, and radiation therapy. Over the last decade, targeted therapies like imatinib (Gleevec®) and trastuzumab (Herceptin®)—drugs that target cancer cells by homing in on specific molecular changes seen primarily in those cells—have also emerged as standard treatments for a number of cancers. And now, excitement is growing for immunotherapy—therapies that harness the power of a patient’s immune system to combat their disease, or what some in the research community are calling the “fifth pillar” of cancer treatment.

One approach to immunotherapy involves engineering patients’ own immune cells to recognize and attack their tumors. And although this approach, called adoptive cell transfer (ACT), has been restricted to small clinical trials so far, treatments using these engineered immune cells have generated some remarkable responses in patients with advanced cancer.

For example, in several early-stage trials testing ACT in patients with advanced acute lymphoblastic leukemia (ALL) who had few if any remaining treatment options, many patients’ cancers have disappeared entirely. Several of these patients have remained cancer free for extended periods. Equally promising results have been reported in several small trials involving patients with lymphoma.

These are small clinical trials, their lead investigators cautioned, and much more research is needed. But the results from the trials performed thus far “are proof of principle that we can successfully alter patients’ T cells so that they attack their cancer cells,” said one of the trial's leaders, Renier J. Brentjens, M.D., Ph.D., of Memorial Sloan Kettering Cancer Center (MSKCC) in New York.

“A Living Drug”

Adoptive cell transfer is like “giving patients a living drug,” continued Dr. Brentjens. That’s because ACT’s building blocks are T cells, a type of immune cell collected from the patient’s own blood. After collection, the T cells are genetically engineered to produce special receptors on their surface called chimeric antigen receptors (CARs). CARs are proteins that allow the T cells to recognize a specific protein (antigen) on tumor cells. These engineered CAR T cells are then grown in the laboratory until they number in the billions. 


The expanded population of CAR T cells is then infused into the patient. After the infusion, if all goes as planned, the T cells multiply in the patient’s body and, with guidance from their engineered receptor, recognize and kill cancer cells that harbor the antigen on their surfaces. CAR T-cell therapy eventually may “become a standard therapy for some B-cell malignancies” like ALL and chronic lymphocytic leukemia, Dr. Rosenberg wrote in a Nature Reviews Clinical Oncology article.

Wish you all a happy, healthy and prosperous 2015! Let us hope 2015 will be a year when reason would prevail over superstitions, light of knowledge over the darkness of blind faith and peace of understanding over the conflicts from misguided convictions! The above short collection is from various sources, special appreciation to Kurzweil's Newsletter, Scientific American and MIT Technology Reviews. 

Science is the best tool ever devised
For understanding how the world works
[Jacob Bronowski]