Friday, September 29, 2017

CRISPR: a case for revision



            It started out as kids; along for the ride as our patents watched science fiction thrillers. The writers and directors would look into the future and ‘wow’ audiences with what they thought it would hold. Star Trek gave us teleportation, Back to the Future gave us time travel, Blade Runner let us look into a world of replicants (artificial intelligence), and many more. What all of these movies had in common though are dreams. As adults now, we have watched some of those dreams materialize, and we’ve watched others merely fade away.

            Accurate and precise gene editing was once one of those dreams. CRISPR was once one of those dreams! Now it is anything but that, and as with every great discovery there is excitement, but there are also many ethical questions that accompany the use of such a tool.

            Clustered Regularly Interspaced Short Palindromic Repeats, more commonly known as CRISPR or CRISPR-Cas9, (once part of a E. Coli defense system) are RNA-guided engineered nucleases. That’s a lot to take in from one definition so I will try to explain it a little better. CRISPR is composed of two subunits: the first subunit is the Cas9 nuclease, which acts like scissors to cut the DNA, and second subunit is a small RNA molecule that can precisely and accurately direct Cas9 to where it is supposed to cut the DNA. Once cut, the new DNA or gene can be incorporated by the repair enzymes. Scientist were able to take advantage of what was originally a defensive mechanism and turn it into an offensive mechanism. (1)

            Wait! Haven’t scientist been able to edit genes for quite some time? The answer is yes, but to a large extent we’ve used applications that are inferior to CRISPR (since its discovery). Targeting the genome before CRISPR was mainly done through engineering DNA nucleases. Two examples of this would be (the oldest and most studied) Zinc Finger Nucleases (ZFNs), and Transcription Activation-Like Effector Nucleases (TALENs). Although both ZFNs and TALENs have been essential for research, their shortcomings for gene therapeutics are made aware by the advantages of CRISPR. ZFNs and TALENs can be expensive, they can be very hard to make, they can be toxic, and they often times (ZFN more so that TALEN) have poor targeting. (2)

            Ben Parker once said, “With great power comes great responsibility” and that is very true in the world of science whenever a new tool like CRISPR is discovered.

            The ability to edit a genome is amazing! It’s exciting, and above all else it has the real power to effect lives. Diseases like malaria, that shake entire continents could be eradicated. HIV, Huntington’s Disease, and any other genetic disease could be a thing of the past. Isn’t that everyone’s goal when choosing a career in healthcare? To help patients, or develop ways to help patients?

            The power to effect change at all is intoxicating, but can sometimes be detrimental. When looking at CRISPR, the ethical questions raised by it are not different from the same ones raised about gene editing/therapy. Most of the ethical problems associated with CRISPR are from a misuse of the tool, but some are purely associated to using the tool at all.
            The first question that comes to mind is for me is the regulation of CRISPR not only in terms of when to use it, but also how it is consumed. I hate to bring up the past, but in a way CRISPR can be seen as a form of positive eugenics. Especially if strict regulations are not placed on it. Theoretically parents in the future could pick and choose how their babies would be edited, leading to “designer” babies. If CRISPR isn’t available to all, or can only be used by those with the resources then that could lead to selective advantages for some (this is all highly hypothetical though). It will take a lot of work from organizations around the world to not only work together for uniform regulation, but to also enforce these regulations across borders. (3)

Also, are the benefits of CRISPR worth the risk? Anytime you work with editing the genome there is a chance that you don’t get your desired effect. There are off-target effects that can be deleterious too if CRISPR is used. If you can cure one disease, but that cure leads to other problems is it worth it? (4)

All of that aside, CRISPR was, is, and will continue to be a discovery for the ages. The very fact that we can have these conversations now is amazing. Elon Musk recently said in an interview with Neil deGrasse Tyson on StarTalk Radio that there were five things he thought would change the future of humanity. They are the internet, sustainable energy, space exploration, artificial intelligence, and rewriting human genetics. All of those are honorable quest, but as future healthcare professionals rewriting the human genome is right up our alley and CRISPR just might be the tool that helps us reach that goal. (5)

             
          


Citations:
1.     Ledford H. CRISPR: gene editing is just the beginning. Nature. 531, 156-159. 10 March 2016.
2.     Mestrovic T. How Does CRISPR Compare to Other Gene-Editing Techniques? News Medical Life Sciences. 13 January 2016.
3.     Mulvilhill J. et al. Ethical issues of CRISPR technology and gene editing through the lens of solidarity. British Medical Bulletin, Volume 122, Issue 1, 1 June 2017, Pages 17-29.
4.     Rodriguez E (2016). Ethical Issues in Genome Editing using Crispr/Cas9 System. J Clin Res Bioeth 7:266. doi:10.4172/2155-9627.1000266
5.     Zimmer C. Breakthrough DNA editing born of bacteria. Quanta Magazine 2015. https://www.quantamagazine.org/20150206-crispr-dna-editor-bacteria/
6.     Welsh J. 5 things Elon Musk believed would change the future of humanity in 1995. Science. Business Insider. 6 April 2015.

 By Christopher Adams, Master's of Medical Science Student, University of Kentucky
           


A class blog on opioids and the treatment of pain


   
Image result for opioid epidemic 2017

The opioid epidemic is thought to be due in part to the over-prescription of drugs like OxyContin to treat patients with symptoms of chronic pain.  These drugs are known as a class as "opioids" that all act via their interactions with the opioid receptors.  By understanding how these drugs work, we can develop new drugs that can still be effective in treating pain, but lack the addictive properties of our currently used drugs. We can also learn more about pain and chronic pain to develop drug-free strategies that lessen the symptoms of pain. 
                How do Opioids cause their effects?   Opioids target opioid receptors within the body in a variety of locations, the central nervous system, peripheral nervous system as well as the gastrointestinal tract. The three original major subtypes of opioid receptors are mu, kappa, and delta opioid receptors within the body. The mu opioid receptor has three subtypes, µ1, µ2, µ3 and is expressed in the brain spinal cord, intestinal tract, and sensory neurons. The kappa opioid receptor also has three receptor subtypes, κ1 κ2 κ3, and is expressed in the brain and the spinal cord. The delta opioid δ1 δ2 receptor is expressed in the brain and sensory neurons. Opioid receptors are G protein coupled receptors that use opioids as their ligands and form hetero and homotrimeric complexes that signal to kinase cascades and aid in the integrity of a variety of proteins. Once the opioid is bound to the active site the receptor undergoes a conformational change, which activates the intracellular G proteins. The α subunit of the G-protein exchanges its bound GDP molecule for an intracellular GTP molecule. This allows the α-GTP complex to dissociate from the βγ complex. These complexes are now able to interact with target proteins. Typically the opioid agonist binds to its G-protein receptor, which results in the inhibition of adenylyl cyclase. This causes a reduction in intracellular cyclic adenosine monophosphate (cAMP) levels. These complexes also interact with a number of ion channels, producing activation of potassium and inhibition of calcium. The net effect of these occurrences cause a reduction of neurotransmitter release, which decreases the generation of the postsynaptic impulse which inhibits tonic neuronal activity. Opioid receptors can also be activated by endogenous peptides that are released by neurons in the brain. There are three pro-hormone precursors that provide the parent compounds these endogenous ligands. Proenkephalin, is cleaved to form met-enkephalin and leu-enkephalin, and bind to the DOP (delta opioid receptor). Prodynorphin which is cleaved to form Dynorphin A and B, these are agonists at the KOP (kappa opioid receptor). β- endorphin is cleaved to form Pro-opiomelancortin (POMC), which is an agonist at the MOP (mu opioid receptor). However, POMC is capable of displaying agonist activity at all three classical opioid receptors.
Schumacher MA, Basbaum AI, Naidu RK. Opioid Agonists & Antagonists. In: Katzung BG, Trevor AJ. eds. Basic & Clinical Pharmacology, 13e New York, NY: McGraw-Hill; 2015.
Sobczak, Marta, et al. “Physiology, Signaling, and Pharmacology of Opioid Receptors and Their Ligands in the Gastrointestinal Tract: Current Concepts and Future Perspectives.”NCBI, US National Library of Medicine, Feb. 2013, www.ncbi.nlm.nih.gov/pmc/articles/PMC3895212/.
               Biased agonism and tolerance.  Opiods exert other actions that proceed via events known as "biased agonism" and "tolerance".  These two events are related.  Biased agonism occurs when a receptor has affinity for multiple ligands. Depending on the ligand, a different signal transduction pathway can be activated, but the ligand specifies which will be activated. Tolerance is a reduced reaction to a drug after repeated use. When a patient is prescribed continuous use of an opiate, tolerance can become a problem in a clinical setting. Since opioids cause analgesic side effects, the dose may be increased at times when the analgesic side effects are no longer as strong. Opioid tolerance associated with biased agonism is currently not a completely understood process, but ongoing research aims to understand the effects. Some scientists argue that an opioid such as morphine may bind a receptor, and even though the morphine is never internalized, the cell is still signaling and causes a tolerance buildup to occur. Other ligands in the body have the ability to become phosphorylated, which changes their structure, and then are internalized and desensitized in the cell, which leads to tolerance. Structurally different agonists for a receptor such as mu can produce different binding efficiencies, which can influence the rate of arrestin recruitment or G protein activation. Research is still being conducted in order to obtain more clear results. Many researchers also hypothesize that because mu receptors are recycled, tolerance is increased.
 Williams, John T. et al. “Regulation of µ-Opioid Receptors: Desensitization, Phosphorylation, Internalization, and Tolerance.” Ed. Annette C. Dolphin. Pharmacological Reviews 65.1 (2013): 223–254. PMC. Web. 18 Sept. 2017.
Al-Hasani, Ream, and Michael R. Bruchas. “Molecular Mechanisms of Opioid Receptor-Dependent Signaling and Behavior.” Anesthesiology 115.6 (2011): 1363–1381. PMC. Web. 18 Sept. 2017.
Anderson, B. (2011). A Pharmacological Primer of Biased Agonism. Endocrine, Metabolic and Immune Disorders: Drug Targets. doi:10.2174/187153011795564179
               Other approaches that can be used to treat pain.  Drugs that can be administered include non steroidal anti-inflammatories, such as acetaminophen, antidepressants, anticonvulsants, muscle relaxants, the use of nerve block procedures (epidural steroid injections; facet joint injections; lumbar sympathetic block) and natural alternatives (arnica gel; aromatherapy; vitamin C).  These drugs and approaches vary with respect to their effectiveness and limitations.  Nonsteroidal anti-inflammatory drugs, like ibuprofen, can be a convenient and effective method of treating pain. They can be bought over-the-counter and are unlikely to cause addiction. They often work well in treating acute pain, such as headaches and muscle aches.  They do, however, have some potentially dangerous side effects when taken for a prolonged period of time. Their metabolism can lead to damage of the liver and/or kidneys. They may also negatively impact the heart, blood clotting, the GI tract, and increase the patient's risk of ulcers. Taking an excessive amount of acetaminophen in 24 hours can cause liver failure; some opioids contain acetaminophen.
               Muscle relaxants can also be used to treat some kinds of muscle pains, however these medications often cause drowsiness. Nerve block procedures are very effective in treating pain, but they must be administered  via injection and must be given by a physician. Tricyclic antidepressants can also be effective in treating chronic pain. The dose of medication needed to treat pain is typically lower than of that required to treat depression. One limitation of these drugs is that the medication must be taken every day, even you aren't experiencing pain a given day. Generally, they should be taken at night because sedation is a side effect. Anti-seizure medications can be taken to treat some nerve pain. This is another medication that is not intended to be taken on an "as needed" basis- and therefore must be taken every day, regardless of the patient's pain level on a given day. Older anticonvulsant drugs required that users have liver activity monitor while taking the medication; the newer drugs do not require liver monitoring, but can still pose a threat to the kidneys. Another limitation would be the type of pain anti-seizure medications can treat- they are suitable for burning and shooting pains.
               Natural alternative are certainly an option and should be considered (the less medication we have to take, the better). They may work surprisingly well for mild pain, but these alternatives are likely to be limited in their effectiveness in treating severe pain. Arnica gel can be used for bruising and muscle soreness. Some users have said the arnica gel helped control their arthritis on a level comparable to ibuprofen. Sufferers of chronic pain like fibromyalgia could consider aromatherapy- for example, massaging essential oils into areas of discomfort. Lastly, it is thought that taking vitamin C supplements may sooth sufferers of some chronic pain, like osteoarthritis.
Wolkerstorfer, A., Handler, N., & Buschmann, H. (2016). New approaches to treating pain. Bioorganic & Medicinal Chemistry Letters,26(4), 1103-1119. Retrieved September 18, 2017, from http://www.sciencedirect.com/science/article/pii/S0960894X15304169
Pain, R. (2000). Limitations of NSAIDs for pain management: Toxicity or lack of efficacy? The Journal of Pain,1(3), 14-18. Retrieved September 18, 2017, from http://www.jpain.org/article/S1526-5900(00)09397-4/fulltext
Dean, L. (2011). Comparing NSAIDs. Pubmed: Clinical Q&A. Retrieved September 18, 2017, from https://www.ncbi.nlm.nih.gov/books/NBK45590/.
Munir, M., MD, Enany, N., MD, & Zhang, J., MD. (2007). Nonopioid Analgesics. Medical Clinics of North America,91(1), 97-111. Retrieved September 18, 2017, from http://www.sciencedirect.com/science/article/pii/S0025712506001179?via%3Dihub
Lukatz, T. (2000). Anticonvulsants for neuropathic pain syndromes: mechanisms of action and place in therapy. Drugs,60(5). Retrieved September 18, 2017, from https://www.ncbi.nlm.nih.gov/pubmed/11129121.
Wiffen, P. (2013). Antiepileptic drugs to treat neuropathic pain or fibromyalgia an overview of Cochrane reviews. Cochrane. doi:10.1002/14651858.CD010567.pub2
Development of new drugs that target the opioid receptor.  Researchers at Tulane University have developed a new painkiller drug that has a comparable strength to morphine but has fewer side effects and likelihood of addiction.  Here, several different endomorphin analogs, referred to as EM analogs, were developed using an endogenous cyclized δ–amino acid-containing endomorphin analog.  Of the four different synthesized EM analogs, the study found EM analog 4 to be the most effective. The engineered EM analogs are highly selective for µ receptors (MOPs), which are the most effective opioid analgesic targets.  The EM analogs acted through binding to the µ receptor, and further decreasing glial P38/CGRP/P2X7 receptor signaling. Activation of this receptor signaling pathway has implications in causing chronic pain.
The study was performed as follows.  First four different EM analogs were synthesized.  Then,  the activation of MOP, DOP, KOP was ensured through completing receptor binding assays that utilized cloned CHO-K1 membranes as the membrane for the respective receptors to function within. To ensure the activation of these receptors by the EM analogs, activation of the receptors was measured through completion of GTPgS assays, which demonstrated over a 95% efficacy for successful MOP activation. Once it was established that the opioid receptors were activated with the EM analogs, trials progressed to an in vivo setting.
               This study used both male CD-1 mice and Sprague-Dawley rats that were exposed to a 12-hour light photoperiod/12-hour dark photoperiod. Drug administration for the Sprague-Dawley rats used an indwelling jugular vein catheter, whereas drug administration for the CD-1 mice was done either through SQ injections in the neck region or through oral routes using a gavage. The same measures were completed for both rats/mice injected with morphine as well as mice/rats injected with EM analogs.
Antinociception was tested through the “tail-flick” test, where rodents were exposed to a heat source at their tail. Reaction latency was recorded with a nine second cutoff time to prevent possible tissue damage. The duration of antinociception for IV, SQ, and oral administration of EM analog 4 were all higher than the respective durations of antinociception for morphine administration.
Respiratory depression was measured in non-anesthetized rats that were allowed free movement within a plethysmography system. Minute ventilation was monitored over a 20-minute time span before drug administration, then again ever 20 minutes after drug administration until antinociception levels had decreased to >50%. Compared to morphine, rodents who were exposed to EM analogs had a decreased level of respiratory depression, and even complete absence of respiratory depression in some rodents.
Motor coordination was examined using a rotorod system, which is a system that measures the ability of an animal to remain upright while on a moving rod. Rodents were given their respective drugs every twenty minutes until a %MPE of greater than 90% was achieved. From there the rats were left on the rotorods for 3 minutes until being removed. Motor coordination was calculated using the Rotomex-5 instrument. Compared to rodents exposed to morphine, rodents who were exposed to EM analogs did not have statistically significant motor coordination impairment whereas their morphine rodent counterparts did. 
Both hyperalgesia and tolerance were tested over a seven-day period, and ED50s values (effective dose values) were compared before and after the seven-day period. Rodents exposed to EM analogs demonstrated less tolerance than the morphine exposed rodents after a seven-day period. EM analog 4, when compared with morphine, demonstrated no induced hyperalgesia whereas the rodents given morphine showed induced hyperalgesia after the seven-day period.
The activation of glial P38/CGRP/P2X7 receptor signaling was measured through using markers for astrocytes, microglia, and MAP kinase and then examined for the expression of the different markers within post-mortem rodents. The rodents injected with morphine demonstrated a greater activation of cell receptor signaling than the rodents injected with EM analogs, which indicates a reduced expression of chronic pain within the EM analog rodents than the morphine rodents.
The risk of dependency and abuse were measured in two different formats, both in conditioned place preference settings and in self-administration settings. For the conditioned place preference setting, rodents could freely roam in a two-compartment box for two days. After two days, the rodents were given their respective opioid drug and confined to only one of the compartments for a 30-minute duration. This cycle of drug administration and 30-minute confinement was repeated for three consecutive days. On the fourth day, the rodents were placed back in the two-compartment box without being given their opioid drug and were allowed free roaming between either compartment. The amount of time spent in each compartment was recorded over a twenty-minute time span. The rodents that had been injected with morphine spent a significantly greater amount of time in the compartment in which they had been administered morphine in rather than the other compartment. The rodents that had been injected with EM analog, however, and demonstrated no preference between the two compartments. For the self-administration setting, rodents were placed in a compartment that had an “active” bar that when pushed on administered their respective drug intravenously, and an “inactive” bar that when pressed did not cause drug release. Infusion release was regulated to only allow a fixed ratio of drug administration over a seven-day period with exposure to the different bars for 12 hours per day. The rodents that received morphine exhibited an increase “active” bar pressing by a five-fold increase. The rodents that received EM analog after pressing their “active” bar did not demonstrate a more frequent bar pressing over the seven-day period.
               One of the limitations of this study was the limited comparison to morphine exposure and not any of the other available opioid drugs. While morphine is a commonly used opioid both in the medical profession and in illegal-use, morphine is not the only drug used in both settings. This study also only compared the effects of morphine and the EM analogs in two rodent species. Another limitation of this study is the duration of time that these measurements were taken over, which fail to demonstrate the same side effects of their drug usage over a longer time interval. Many opioid drugs demonstrate greater side effects and complications during prolonged use for chronic pain, and an experimental setting that only examined these parameters concerning opioid drug side effects during a seven-day period cannot be used to predict possible implications on the same drug usage over a longer interval.
               While the EM analogs described in this study, particularly EM analog 4, demonstrated an improved analgesic-to-side-effect ratio, several steps need to be taken to determine the possible applications of EM analog 4 in the medical field. Similar studies need to be done that compare other opioid drugs, such as fentanyl and codeine. Similar studies also need to be completed over a larger time span to determine the applications of EM analog 4 in the treatment of chronic pain, seeing that many of the instances that use opioid drugs for the management of chronic pain. Once studies such as these have been completed within the rodent species, comparable experiments need to be conducted using primate species before any human implications can be derived through the substitution of EM analogs for pain management in the medical field.
Zadina, J., Nilges, M., Morgenweck, J., Zhang, X., Hacker, L., & Fasold, M. (2015). Endomorphin analog analgesics with reduced abuse liability, respiratory depression, motor impairment, tolerance, and glial activation relative to morphine. Neuropharmacology,105, 215-227. Retrieved September 18, 2017, from http://www.sciencedirect.com/science/article/pii/S0028390815302203