NIH Director's New Innovator Award Program

NIH Director’s New Innovator Award Program Highlights

Researchers define precise EEG Signature of Anesthesia-Induced Unconsciousness

EEG Signature of Anesthesia-Induced Unconsciousness
More than 60,000 patients in the United States undergo general anesthesia for surgery every day. Currently, doctors use indirect measures of the brain state, such as heart rate and blood pressure, along with drug pharmacokinetics, pharmacodynamics and exhaled anesthetic gas typically are converted to a simple index score to indicate if a patient is adequately anesthetized during surgery. These indices are neither entirely reliable nor very informative. Inadequately administered anesthesia can result in intraoperative awareness and post-operative delirium. Drs. Emery N. Brown and Patrick L. Purdon, former NIH Director’s Pioneer Awardee and current NIH Director’s New Innovator Awardee, respectively, have studied brain activity during propofol-induced anesthesia using electroencephalogram (EEG) technology, which measures scalp electrical potentials, to better understand the mechanism of unconsciousness and to establish a direct method of tracking the transition between consciousness and unconsciousness during general anesthesia. In this study, the researchers gradually administered and reduced the anesthesia drug propofol in patients, while monitoring brain activity and behavioral loss of consciousness by asking patients to respond to auditory stimuli. This gradual induction and emergence from anesthesia allowed the researchers to precisely define an EEG brain signature, as well as behavioral markers that are associated with consciousness, unconsciousness, and the transition between these two states in patients undergoing propofol-induced general anesthesia. This research has provided a deeper understanding of the mechanism of propofol-induced loss of consciousness, and could be used to determine the EEG brain signatures of other anesthetics. Additionally, these results could be used to develop more direct and reliable methods for monitoring brain states of patients undergoing general anesthesia and could lead to new insights into the tailoring of drug dosage in real-time.

Reference:

P. L. Purdon et al., Electroencephalogram signatures of loss and recovery of consciousness from propofol. Proceedings of the National Academy of Sciences, (March 4, 2013, 2013).
 

Up to Top

SUCCESS WITH GENE CIRCUITS COULD LEAD TO NEW APPROACHES TO GENE THERAPY

SUCCESS WITH GENE CIRCUITS COULD LEAD TO NEW APPROACHES TO GENE THERAPY
A gene circuit refers to the complex mechanism by which genetic material, encoded by DNA, interacts with proteins resulting in a specific phenotype, or an observable characteristic. This can be more readily understood by imagining an electrical circuit, where you can turn a light switch on or off to produce an observable characteristic of light or darkness, respectively. Gene circuits can be turned on or off just like a switch, thus the expression of the associated phenotype can therefore be turned off or on. Additionally, these circuits can be expressed as more than just on or off. Imagine a light switch with a dimming function by which the light can be expressed at different intensities. Similarly, these gene circuits can be regulated in order to express at different intensities rather than just on or off, in order to produce phenotypic differences at varying levels of expression. In nature, multiple genes in multiple gene circuits of a cell interact to regulate major biological processes like cell signaling or division. Researchers can construct simple synthetic gene circuits in order to study the expression of specific genes in living systems, such as microorganisms, as well as to create novel biological functions for practical applications.

NIH Director’s New Innovator Awardee, Gábor Balázsi, and colleagues have described a new method where they developed and optimized a synthetic gene circuit in yeast and were able to adapt the gene circuit to mammalian cells. Importantly, the authors optimized this gene circuit allowing them to finely adjust gene expression in a linear fashion with uniform gene expression across the entire cell population. Previously, scientists had been unable to directly transfer gene circuits developed in microorganisms directly into mammalian systems, such as human cells. Dr. Balázsi’s new findings will allow researchers to design circuits that can be tested and characterized in microorganisms, such as yeast, which are easier to engineer than mammalian cells, and then introduce the circuits into mammalian cells, a model more similar and representative to study the complexities of human disease. These gene circuits can be used to study the effects of specific gene expression on biological functions such as development, immune response, and nervous system response, and can be used to develop precisely controlled gene expression systems for use in individualized gene therapy to treat specific diseases and genetic conditions.

Reference:

D. Nevozhay, T. Zal, G. Balazsi, Transferring a synthetic gene circuit from yeast to mammalian cells. Nature communications 4, 1451 (Feb 5, 2013).
 

Up to Top

NEW MODEL ALLOWS SCIENTISTS TO STUDY MECHANISM OF HCM DEVELOPMENT AND TEST DRUGS FOR TREATMENT

Hypertrophic Cardiomyopathy (HCM)
Hypertrophic Cardiomyopathy (HCM) is a prevalent heart condition that involves the thickening of the heart walls, which overtime can lead to the development of an irregular heart beat and sudden cardiac death. HCM has been estimated to be the most common inherited heart condition in the world, and has been associated with several DNA mutations affecting sarcomeres, or the basic unit of a muscle.

Dr. Joseph Wu, an NIH Director’s New Innovator Award Recipient from Stanford University, and his colleagues, have developed a new technology using induced pluripotent stem cells (iPSCs) obtained from skin cells of a family of patients that allows them to model HCM at the level of a single cell. The researchers were able to turn the skin-derived iPSCs into heart muscle cells, permitting them to investigate the underlying causes of HCM development without requiring the difficult direct collection of diseased heart tissue. This model allowed the researchers to better understand how a specific DNA mutation, carried by affected members of this particular family, can lead to the development of HCM, and helped them to test therapies that might be used to treat or prevent HCM due to this specific mutation. This new technology can be used by other researchers to investigate the effect of other DNA mutations on the development of HCM, and to screen the effectiveness of new treatments and preventative therapies specific to each mutation.

Read the university news release... Exit Disclaimer

Read the San Francisco Chronicle Article... Exit Disclaimer

Reference:

Lan F, Lee AS, Liang P, Sanchez-Freire V, Nguyen PK, Wang L, Han L, Yen M, Wang Y, Sun N, Abilez OJ, Hu S, Ebert AD, Navarrete EG, Simmons CS, Wheeler M, Pruitt B, Lewis R, Yamaguchi Y, Ashley EA, Bers DM, Robbins RC, Longaker MT, Wu JC. Abnormal calcium handling properties underlie familial hypertrophic cardiomyopathy pathology in patient-specific induced pluripotent stem cells. Cell Stem Cell. 2013 Jan 3;12(1):101-13. PubMed PMID: 23290139.
 

Up to Top

 

Self Assembling DNA Bricks

Self Assembling DNA Bricks
Dr Peng Yin and Dr. William Shih, both awardees of the Common Fund NIH Director’s New Innovator Award program, have described a new method to construct complex three dimensional structures by using short synthetic DNA strands which the investigators call “DNA bricks.” In this paper, the investigators used 32-nucleotide DNA strands which self-assembled into prescribed 3D structures without scaffolds. Each DNA brick can bind up to four local neighbors, and in a manner similar to Lego® bricks can be used to build complicated structures. This self assembly has allowed the investigators to create over 100 distinct structures which can be found here. Exit Disclaimer This method may have a large variety of applications such as to develop smart drug delivery particles or provide a better way for nano-scale fabrication of inorganic materials.

Reference:

Yonggang Ke, Luvena Ong, William Shih, Peng Yin Three-Dimensional Structures Self-Assembled from DNA Bricks, Science - 30 November 2012 (Vol 338, pp1177-1183) More information describing the self assembly process can be found in the two videos below:

Video 1 Exit Disclaimer Animated video containing narration which describes how 3D structures of DNA bricks are assembled.

Video 2 Exit Disclaimer Animated video which depicts the nanofabrication technique called “DNA-brick self-assembly” using short synthetic strands of DNA that work like interlocking Lego® bricks.

Read more about New Innovators Awards...

 

Up to Top

Stress-Amplifying Protein Linked to Diabetes

Stress-Amplifying Protein Linked to Diabetes
Dr. Feroz Papa, an associate professor of medicine at the University of California San Francisco and NIH Director’s Transformative Research Award recipient discovers a molecule amplifying stress in the earliest stages of diabetes.



 

Up to Top

Tele-medical Device Wins Top Honors

Tele-medical Device
Dr. Aydogan Ozcan, an NIH Director’s New Innovator Award recipient, and colleagues at the University of California, Los Angeles (UCLA) garner first place in The Scientist magazine, “Top Ten Innovations of 2011” competition by developing a cell phone camera attachment that functions as a mobile microscope.

The device known as LUCAS (Lensless, Ultra-wide-field Cell monitoring Array platform based on Shadow imaging), is a light-weight holographic microscope that uses inexpensive off-the-shelf parts as an attachment to a cell phone’s camera. Biospeciman samples are loaded into the slide attachment of the device, and then battery operated light emitting diodes (LEDs) are used to illuminate the specimen and produce an image of the cells shadows. The image is then remastered using an algorithm remotely run from a computer. Although the resolution of the LUCAS is at the submicrometer level, the microscope can be used for wide-field imaging.

The technology coupled with the use of a cell phone provides a plethora of opportunities to capture and electronically transport biospeciman images in real time. The device can be used for water quality testing and even to monitor diseases such as Malaria. At the estimated cost of approximately 10 USD for the LUCAS attachment, the technology has the potential of providing diagnostic services to resource limited areas across the nation. Field tests around the world are already being planned for the LUCAS in 2012.

Reference:

Mudanyali O, Tseng D, Oh C, Isikman SO, Sencan I, Bishara W, Oztoprak C, Seo S, Khademhosseini B, Ozcan A. Compact, Light-weight and Cost-effective Microscope based on Lensless Incoherent Holography for Telemedicine Applications, 2010.June10. PMCID: PMC2902728
 

Up to Top
New material holds promise for drug delivery, medical implants

New material holds promise for drug delivery, medical implants

Dr. Adah Almutairi, a 2009 NIH Director’s New Innovator awardee, and colleagues at the University of California San Diego, have developed a new type of “smart” polymeric material that may have widespread medical and biological applications. Reported in the journal Macromolecules, the new material disassembles in response to harmless levels of near infrared (NIR) irradiation, which can penetrate up to 10 centimeters (almost 4 inches) into the body. Both the material itself and its breakdown products are well-tolerated by living cells, suggesting this material would potentially be safe for use in humans. This type of material could be used as a capsule for a drug, allowing doctors to only release the drug in a specific area, such as right next to a tumor. It could also be used in tissue engineering, implants, wound-healing, and biosensors. Dr. Almutairi and colleagues are now working on improving the design of this polymeric material, so that it becomes even more sensitive to NIR, allowing a more controlled disassembly of the material. This research is a significant step forward in the development of light-sensitive materials that will allow doctors and researchers to target previously inaccessible sites with precise spatial and temporal control.

References:

Fomina N, McFearin CL, Sermsakdi M, Morachis JM, and Almutairi A. Low power, biologically benign NIR light triggers polymer disassembly. Macromolecules, 2011. 44: 8590-7. PMID: 22096258.


 

Up to Top
Brain stem cell environment

Location, location, location: Scientists uncover new information about brain stem cell environment

The adult mammalian brain contains several specialized areas where stem cells capable of producing new neurons reside. Dr. Chay Kuo, an NIH Director’s New Innovator Award recipient, has identified key components of one such specialized area, or niche, that is critical for the production of new neurons. Published in the July 14, 2011 issue of Neuron, Dr. Kuo and colleagues demonstrate that two proteins, Foxj1 and Ank3, are critical for maintaining the cellular niche around brain stem cells.

Foxj1 is a transcription factor, a type of protein that regulates when and where other genes are expressed. Within the stem cell niche studied by Dr. Kuo, the Foxj1 protein causes the Ank3 protein to be expressed. The Ank3 protein then helps assemble groups of ependymal cells, a type of cell that surrounds the brain stem cells and provides support for the production of new neurons. Without Foxj1 and Ank3 proteins, the niche is disrupted and stem cells fail to produce new neurons.

Currently, when neural stem cells are studied in the laboratory, they are not surrounded by these ependymal cells. Under these conditions, it is extremely difficult to make the stem cells produce neurons. The findings of Dr. Kuo and colleagues suggests that in the laboratory setting, scientists may need to recreate the same kind of support provided by ependymal cells in the brain stem cell niche. By understanding the local cellular environment that helps support production of new neurons, researchers hope to improve future therapeutic strategies that use stem cells to produce neurons for repairing or replacing damaged tissue.

References:

Paez-Gonzalez P, Abdi K, Luciano D, Liu Y, Soriano-Navarro M, Rawlins E, Bennett V, Garcia-Verdugo JM, and Kuo CT. Ank3-dependent SVZ niche assembly is required for the continued production of new neurons. Neuron, July 14, 2011. 71: 61-75. PMID: 21745638.


 

Up to Top
High Throughput

High Throughput Strategy for Testing Nerve Regeneration

Finding new drugs to promote regeneration of damaged nerve cells holds great promise for diseases such as Alzheimer's disease, spinal cord injury, brain trauma, and more. Many potential treatments, while promising in cell cultures, fail to promote regeneration in living animals. In a paper published October 13th in the Early Edition of the Proceedings of the National Academy of Sciences, Dr. Mehmet Yanik, a researcher at the Massachusetts Institute of Technology and an NIH Director's New Innovator awardee, demonstrates a novel method to rapidly screen potential drugs for their ability to promote nerve regeneration in the nematode C. elegans. Using this method, Dr. Yanik and colleagues discovered that compounds which regulate protein kinase C (PKC), an enzyme important for many different cellular processes, can modulate nerve regeneration after injury in specific neurons. The ability of this method to efficiently screen large numbers of potential drugs in living animals may greatly accelerate the discovery of new treatments to promote nerve regeneration.

Reference:

Samara C, Rohde CB, Gilleland CL, Norton S, Haggarty SJ, Yanik MF. Large-scale in vivo femtosecond laser neurosurgery screen reveals small-molecule enhancer of regeneration. Proceedings of the National Academy of Sciences, 2010 Oct 26; 107(43): 18342-7. Epub 2010 Oct 11. PMID: 20937901.




 

Up to Top
Brain Activity

New Tools to Correct Brain Activity

Many neurological and psychiatric disorders are associated with abnormal activity in specific brain circuits. Historical approaches to correct abnormalities in brain circuits have relied on the use of electrical or magnetic stimulation, which only relieve the symptoms partially or for a short time. Although contemporary electromagnetic stimulation techniques have overcome many of the drawbacks of earlier approaches, a continuing and pressing need remains for new medical approaches to systematically correct abnormal brain function in patients with epilepsy, brain injury, and Parkinson’s disease.

Dr. Ed Boyden in the Common Fund’s New Innovator program has engineered a powerful new class of tools to shut down nerve activity for short periods of time using different colors of light. These techniques are based on genes recovered from bacteria and fungi that encode light-activated proteins normally used for energy production in these organisms. When nerve cells expressing these proteins are exposed to the appropriate wavelength of light, they are prevented from transmitting electrical signals. When used in combination with genetic techniques to target these proteins to specific brain regions or cell subsets, these tools can lead to a much deeper understanding of the brain’s role in health and disease. The development of new technologies that allow precise control of neural circuits could lead to new treatments for disorders associated with abnormal brain activity, including chronic pain, epilepsy, brain injury, and Parkinson’s disease.





 

Up to Top



  • Download Readers:
  • Download Adobe PDF Reader
  • Download Microsoft PowerPoint Viewer
  • Download Microsoft Word Viewer
  • Download Microsoft Excel Viewer
Division of Program Coordination, Planning, and Strategic Initiatives  •  National Institutes of Health  •  Bethesda, Maryland 20892