2017-08-09

[#DIV28SUPER] NIDA Neuroscience Update August 10, 2017: Funding Opportunities Announcements and a Learning Opportunity

Table of Contents:

I.                 Avenir Award Program for Genetics or Epigenetics of Substance Use Disorders (DP1) PAR-16-357

II.                Identification of Genetic and Genomic Variants by Next-Gen Sequencing in Non-human Animal Models (U01) (PAR-15-120)

III.              Pharmacogenomics of Anti-retroviral Therapy in People Who Inject Drugs (R01)  (RFA-DA-18-014)

IV.            HIV-associated neuropathic pain and opioid interaction (R01)  (RFA-DA-18-015)

V.              Short Course on the Genetics of Addiction at The Jackson Laboratory in Bar Harbor, Maine September 10-16, 2017 www.jax.org/addiction

 

I.                 Avenir Award Program for Genetics or Epigenetics of Substance Use Disorders (DP1) PAR-16-357

Application Receipt Date(s): October 19, 2017; October 19, 2018, by 5:00 PM local time of applicant organization

 

Funding Opportunity Purpose

Avenir means future in French, and this award looks toward the future by supporting early stage investigators proposing highly innovative studies. The award will support those in an early stage of their career who may lack the preliminary data required for an R01 grant, but who propose high impact research and who show promise of being tomorrow's leaders in the field. NIDA has developed two Avenir Award Programs, one for HIV/AIDS research and the other for genetics or epigenetics studies.

 

The Genetics or Epigenetics of Substance Use Disorders Avenir Award program supports early stage investigators proposing highly innovative studies that open new areas of research for the genetics or epigenetics of addiction.  These may be novel methods or approaches that can potentially be applied to the analysis of the genetics or epigenetics of addiction.  Investigators outside the field of addiction interested in applying their novel approaches to the genetics or epigenetics of addiction are encouraged to apply.  The award will support those in an early stage of their career who may lack the preliminary data required for an R01 grant, but who propose high impact research and who show promise of being tomorrow's leaders in the field of genetics or epigenetics of substance use disorders.   

 

 

II.               Identification of Genetic and Genomic Variants by Next-Gen Sequencing in Non-human Animal Models (U01)  (PAR-15-120)

National Institute on Drug Abuse

Application Receipt Date(s):  October 20, 2017; March 1, 2018, by 5:00 PM local time of applicant organization

 

Funding Opportunity Purpose

This announcement encourages applications for projects aimed at the discovery of gene variants in outbred or selectively bred non-human animals through the use of Next-Gen Sequencing technologies. The proposed projects should be based on data demonstrating the relevance of the traits to drug abuse behaviors and processes of addiction.  Investigators may employ previously selectively bred animals, re-derived strains, strains selected for some specific new phenotypes, beginning with a novel progenitor population, or an outbred population. Vulnerability phenotypes, for purposes of this FOA, are defined as individual differences that convey increased propensity to acquire, maintain or escalate to uncontrollable, compulsive drug intake, or increased vulnerability to relapse to drug seeking and drug-taking following a period of abstinence.  Vulnerability phenotypes may be defined behaviorally or neurobiologically, must have demonstrated heritability, and be suitable for mapping in outbred or selectively bred strains. The following are examples of vulnerability phenotypes that have been characterized behaviorally which would be appropriate for study; however, this is not an inclusive listing and there may be others:

 

High drug sensitivity, reactivity or preference

Preference or sensitivity for non-drug rewards

Somatic and affective drug withdrawal

Novelty preference or novelty seeking

Increased incentive motivation for reward-related stimuli

Sensitivity to develop escalation of drug taking

Impulsivity

Poor cognitive flexibility (e.g., reversal learning, set shifting, etc.)

Resistance to punishment during drug-seeking

Persistent responding in the absence of drug

Heightened relapse and reinstatement

Enhanced stress reactivity

Disrupted circadian rhythms

 

 

III.             Pharmacogenomics of Anti-retroviral Therapy in People Who Inject Drugs (R01) (RFA-DA-18-014)
National Institute on Drug Abuse
Application Receipt Date(s): December 14, 2017, by 5:00 PM local time of applicant organization.

 

Funding Opportunity Purpose

Antiretroviral therapies have been effective in treating HIV infection. However, variable responses to anti-retroviral therapy result from genetic variants affecting drug disposition, drug penetration, drug metabolism, drug interactions with drugs of abuse, off-site drug targets and adherence. Induction of enzymes and drug transporters in active drug users and those that relapse may affect treatment efficacy.   The identification of genetic variants that affect HIV treatment toxicity such as CNS side effects, liver and renal failure has broad implication for improving treatment outcomes for the treatment of HIV in people who inject drugs.  In addition predicting which drug regimen will ensure greatest recovery of CD4 function and prevent cognitive decline.  This initiative will use genome sequencing strategies and genome wide association methods to identify genetic variants affecting the pharmacokinetics, pharmacodynamics, and HIV treatment toxicities in people who inject drugs. The research questions to be answered by this initiative areHow do genetic variants affect the response to anti-retroviral therapy in people who are injecting drugs?

  • How does injection drug use affect the pharmacokinetics and pharmacodynamics of anti-retrovirals in people who inject drugs both during active use and during abstinence?
  • What genetic and epigenetic factors predict the greatest recovery of CD4+ function and prevent cognitive decline in injecting drug users in response to antiretroviral HIV therapy?
  • How does impulsivity such as measured by delayed discounting affect compliance to anti-retroviral therapy?  Are there genetic or epigenetic variants that are associated with delayed discounting that impact compliance?

IV.            HIV-associated neuropathic pain and opioid interaction (R01)  (RFA-DA-18-015)
National Institute on Drug Abuse
Application Receipt Date(s): December 18, 2017, by 5:00 PM local time of applicant organization.

 

Funding Opportunity Purpose

In this FOA, NIDA seeks to promote research investigating the underlying mechanisms by which opioids, including prescription drugs, exacerbate HIV-associated neuropathic pain. Results from these studies may help obtain information for the development of safe and effective treatments of neuropathic pain for HIV-infected patients exposed to opioids.

 

Note: The choice of biological system for the proposed investigations should be well justified. Applicants may propose studies that investigate body fluids or cells/tissues from human patients or non-human primates. Other models may be used (e.g. primary cells, cell lines, or animal models), but applicants should provide a strong justification for their choice of model system. Also a rationale should be developed for why these studies will provide foundational knowledge to eventually develop safe and effective treatments of neuropathic pain for HIV-infected patients exposed to opioids.      

 

Applications proposing drug development or NIH-defined clinical trials will not be accepted and will be withdrawn without review.

 

Topics appropriate for this FOA include but not are limited to the following:

 

The following studies are suggested for investigating the underlying mechanisms by which opioids exacerbate HIV-associated neuropathic pain.

 

Studies designed to understand the role of ion channels such as sodium and calcium  

Studies dissecting the relative roles of glutamatergic and GABAergic signaling

Studies of the role of glia cells (microglia and astrocytes) and perivascular macrophages or neuronal-glial cross talk

Evaluations of the role of inflammatory cytokines and chemokines

Investigations of the role of TLR4 signaling

Studies of the contributory role of gut-derived bacterial endotoxin

Studies dissecting interacting effects of HIV, Opioids, and ART

Investigating the impact of genetic variants of opioid receptors

            Investigating potential epigenetic mechanisms     

 

 

V.              Short Course on the Genetics of Addiction at The Jackson Laboratory in Bar Harbor, Maine September 10-16, 2017 www.jax.org/addiction

 

Registration scholarships are availableGo to scholarship link for more details on awards.Registration Fee: $1500 or reduced rate with Scholarship Award.
Fee includes all meals and lodging at The Highseas Conference Center.

Event Contact: Erin McDevitt; erin.mcdevitt@jax.org or 207-288-6659

 

This JAX short course brings together world-renowned experts in addiction, human genetics, and mouse genetics. Through a combination of lectures and hands-on computational modules, the course will feature:

 

·        New methods and applications of mouse genetics to addiction,

·        Genetic and bioinformatics approaches to augment behavioral studies, and

·        Techniques for analyzing human genetic studies of addiction.

 

The course also provides key opportunities to network with students, researchers, and other professionals; and explore potential scientific collaborations.

 

Genetics of Addiction is geared toward students of all experience levels, from undergraduate and graduate students who seek an introduction to the field, to experienced addiction researchers who wish to hone their genetic skills and knowledge. Course attendees are invited to bring their own data for analysis during the hands-on laboratory sessions.

 

Don’t miss this unique opportunity to learn the latest tools and approaches in addiction and genetic research, allowing students and researchers alike to return to their own communities and make meaningful contributions to science and society.

 

This course is supported by a grant from the National Institutes on Drug Abuse under Award Number R13DA032192

 

The National Institute on Drug Abuse at the National Institutes of Health is an agency of the United States Department of Health and Human Services  TO UNSUBSCRIBE: send email to listserv@list.nih.gov Copy and paste UNSUBSCRIBE NIDA_NEURO_SCIENCE-L   in the message body of the email - You will receive a confirmation email if successful. If you have problems contact jpollock@mail.nih.gov   301-435-1309

 

 

No comments:

Post a Comment