BioQuakes

AP Biology class blog for discussing current research in Biology

Tag: human gene editing

A Potential Solution to the HIV Disease with CRISPR?

We always hear about how STDs like HIV can be fatal yet not curable. However, recent advances and research regarding CRISPR (which stands for Clustered Regularly Interspaced Short Palindromic Repeats) has shown that there might be a potential solution for HIV’s. 

 

The Human immunodeficiency virus (HIVs) is an infection that attacks the human body’s immune system. Specifically, they attack the body’s white blood cells, which weakens the immune system and make humans more likely to get sick with some diseases like tuberculosis, infections and some cancers.

 

As of today, the technology and medicine developments allows those who are infected with HIV to take certain medicines that stops the virus from reproducing. As long as patients take the medicine everyday, the medicine provides a temporary remedy. 

 

However, the issue is that when people are first infected with HIV, the HIV viruses can insert their DNA into human’s immune cells, where they stay dormant. So when those that are infected with HIVs stop taking the medicine, the virus can “awaken” and start attacking the immune system again. Thus it is clear that the medicine cannot be a long term solution. 

 

Thanks to the CRISPR technology, there might be a potential solution to this issue. CRISPR is a gene editing technique that edits or deletes a specific part of a gene sequence, which has the ability to disable certain viruses. In this system, a DNA cutting protein called CAS-9 and the guide RNA molecule promote this process. This complex can locate specific locations in a gene sequence and CAS-9 can edit or delete that segment. 

 

While CRISPR is designed to be highly precise, there are still some risks associated with mutations for the technology. In CRISPR, the guide RNA is used to identify the specific sequences; however, there is the risk that it will identify a sequence that is similar to the target sequence and make unintended edits to the particular sequence. This can lead to undesired mutations that could have serious implications. In class, we learned about the different types of mutations that could take place, including silent, missense, nonsense, as well as frameshift. If the mutation happens to be a nonsense or frameshift mutation, it could cause serious implications as the large parts of the gene will either be not read at all, or it will be translated into completely unintended proteins. Nonetheless, despite the risks associated, scientists are now working to perfect the technology. 

As of now, the CRISPR technique has already been approved last year in the US and UK as a treatment for sickle cell anemia. For HIV, there has been research that shows that CRISPR could disable viruses in immune cells, making large progress. Although using CRISPR for HIV is relatively new, scientists have high hopes that this could be a potential solution for the disease. Personally, I think that the CRISPR technology is a revolutionary technology that could be the remedy for many different diseases that are associated with viruses. When taking into account both the revolutionary potentials provided by the CRISPR technology as well as the dangerous risks associated, what are your thoughts on this technology?

 Cancer Detection Using CRISPR Gene Editing

Currently, many are accustomed to invasive cancer diagnostic methods such as endoscopies, colonoscopies, and mammograms. Driven by the desire to discover new methods, a group of researchers from the American Cancer Society developed an alternative method, which is a significant contribution to cancer detection.

Utilizing CRISPR gene editing as their approach, the group of ACS researchers developed an easy-to-use mechanism for detecting small amounts of cancer in plasma. CRISPR gene editing is a method that scientists and researchers have been using to modify an organism’s DNA. CRISPR gene editing is often done for numerous reasons, such as adding or removing genetic material, creating immune defense systems, and repairing DNA. Their detection method also allows healthcare professionals in diagnostics to decipher between malignant and benign cancer-related molecules that they may discover.

CRISPR Gene-Editing

The first step that the researchers made to develop this approach was to design a CRISPR system that creates a manufactured exosome out of two reporter molecule fragments, which they cut. An exosome is a small vesicle that carries material such as lipids, proteins, and nucleic acids after branching out from a host cell. Exosomes are typically involved in detecting cancerous cells because they provide a glimpse into the host cell they branched out from. Therefore, cancerous cells are shown in their exosomes through biomarkers, like micro RNAs (miRNA). In AP Biology class, microRNAs are described as materials that bind to complementary mRNAs to prevent the translation from occurring. MiRNAs are a recent discovery, identified in 1993. It is now concluded that most gene expression is influenced by them, so the researchers made efficient use of miRNA in their experiment. The two fragments of the reporter molecule came together and interacted with the CRISPR’s materials.

Micro RNA Sequence

The researchers concluded that if the targeted miRNA sequence was evident in the combination, the CRISPR system they made would become activated and cut apart the reporter molecule. The researchers specifically targeted miRNA-21, which is often involved in cancer development. The researchers were able to detect miRNA within a combination of similar sequences and later tested their method on a group of healthy exosomes and cancerous exosomes. Their CRISPR system successfully differentiated between the healthy and cancerous exosomes, which makes this system effective for cancer detection. The researchers are confident that their CRISPR gene editing approach to cancer detection will make diagnosis easier on patients and a more efficient process overall.

 

A Vision For a Better Future

CRISPR is a world changing technology that is essentially used to edit genes. The discovery of CRISPR took place in the University of Alicante, Spain. Reported in 1993, Francisco Mojica was the first to characterize CRISPR locus. Throughout the 90s and early 2000s, Mojica realized that what was once reported as unique sets of repeat sequences actually shared common features, which are known to be hallmarks of CRISPR sequences. Through this finding, Mojica was able to correctly hypothesize that CRISPR is an adaptive immune system. In the year 2013, Feng Zhang, was the first scientist to successfully adapt CRISPR-Cas9 for genome editing in Eukaryotic Cells. Zhang was able to engineer two different Cas9 orthologs and he then demonstrated targeted genome cleavage in both human and mouse cells. They discovered that this system could then be used to target multiple genomic loci and could also drive homology directed repair.

CRISPR-Cas9 mode of action.png

How Does it Work?

“Clustered regularly interspaced short palindromic repeats,” also known as CRISPR, are repeats found in bacteria’s DNA. CRISPR-Cas9 was adapted by scientists from a naturally occurring genome editing system in bacteria. This bacteria captures parts of DNA from invading viruses and it uses them to create DNA segments known as CRISPR arrays. This DNA allows the bacteria to recognize and remember the virus’s. If the same virus, or a similar one, attacks again, the bacteria will consequently RNA segments in order to target the viruses DNA. After, the bacteria uses the enzyme Cas9 in order to cut the DNA apart, thus disabling the virus. Scientists in a lab will create small pieces of RNA that attach to a specific target sequence of DNA and also the Cas9 enzyme. In this process, the RNA is used to recognize DNA and the Cas9 will cut the targeted DNA. Once cut, researchers will utilize the cell’s ability to repair DNA in order to add or remove pieces of genetic material. It can also replace existing DNA with custom DNA in order to make changes.

How is it used?

CRISPR is a tool that can be used to fight cancer among other known diseases. The therapy involves making four modifications to T-cells. T-cells are cells that help fight cancer. CRISPR adds a synthetic gene that gives the T-cells a claw-like receptor. This receptor can locate NY-ESO-1 molecules on cancer cells. CRISPR is then used to remove three genes. Two of the removed genes can interfere with the NY-ESO-1 receptor and the third limits a cell’s cancer killing abilities.

Another way CRISPR is used is against Leber’s Congenital Amaurosis(LCA). LCA is a family of congenital retinal dystrophies that results in vision loss. Patients tend to show nystagmus, sluggish pupillary responses, decreased visual acuity and photophobia. The CRISPR trial focuses on one gene mutation that causes a severe form of degeneration. It is said that this mutation creates somewhat of a “stop sign,” and RNAs will target sequences on either part of the stop sign. The Cas9 enzyme will then cut them out, allowing the DNA to then repair itself.

Cell Cycle Regulation in Revolutionary Gene Editing Technique (a.k.a. CRISPR)

There are more than 500 different types of human cancers. Wouldn’t it be wonderful if scientists could develop cures for all of them? Scientists believe that CRISPR gene-editing can be used to cure some cancers. CRISPR (an acronym for clustered regularly interspaced short palindromic repeats) is a way of targeting a specific bit of DNA inside a cell which can then be gene-edited to change such bit of DNA. CRISPR has also been used for other purposes, such as turning genes on or off without changing their DNA sequence.

 

Recent research has found a link between CRISPR gene-editing and mutated cancer cells. Scientists believe that a further understanding of this link can identify a group of genes which should be monitored for mutations when cells are subjected to the CRISPR gene-editing method. Although CRISPR gene-editing holds promise for cell repair, the application of CRISPR gene-editing, which is meant to identify and correct damage in cells, can also cause damage to cells in a controlled manner. Such damage activates a protein, p53 (“also known as the guardian of the genome”), which helps repair damaged DNA. 

CRISPR-Cas9 mode of action

P53 is a transcription factor, which is a protein that regulates the rate at which DNA is transcribed into RNA. These transcription factors bind to regulatory sequences in proteins, thus changing the shape of DNA, ultimately making them the most vital form of gene regulation. Transcription factors include many proteins but exclude RNA polymerase, which pries two strands of DNA apart and joins two strands of DNA together (Campbell, 280). P53 works by sliding along the damaged DNA, seeking a critical site to which it attaches and then sends a message to halt cell division until the DNA is repaired. In other words, p53 acts as a checkpoint in the cell cycle, preventing cell from proceeding though the G1 and G2 phases of the cell division cycle. In mice, the same exact transcription factor exists; those that lacked the Trp53 gene developed tumors at a far faster rate than those with the functioning gene.

 

By using CRISPR technology to damage DNA at the same cite at which DNA damage occurs, scientists are able to identify the protein responsible for cellular proliferation. If damage to the cell is too severe then p53 triggers apoptosis (the death of cells which occurs as a normal and controlled part of an organism’s growth or development) so that the damaged cell is destroyed. However, sometimes p53 is itself damaged which prevents such protein from binding to the damaged DNA in order to repair it or otherwise signaling destruction of the cell. When this occurs, the damaged cells multiply and grow, resulting in tumors. Scientists have found alterations in p53 in more than half of all cancers and thus, consider p53 the most common event in developing cancer.

 

New studies show that p53 inhibition can make CRISPR more effective thus, counteracting “enrichment” (the process of purifying cells for downstream applications such as qRT-PCR, cell polarizations ex vivo, or to enrich cells for use in a flow cytometry experiment) of cells with p53 mutations which has been observed to occur in cell cultures when such cells have been subjected to CRISPR. In other words, there is in vitro evidence that CRISPR technology causes harmful p53 mutations to be more prevalent in the population that has been subjected to the CRISPR technique. These findings suggest that there is a group of genes that should be monitored for mutations when the CRISPR gene-editing method is applied to cells. 

 

Cancer is a devastating disease that has taken the lives of many people. Members of my family have suffered and lost their battle to cancer (most recently my dear aunt this past weekend). CRISPR presents the possibility of finding cures to cancer which are specifically designed to target the particular genetic mutations that are unique to each individual. Perhaps, the cure to cancer will be achieved sooner than we realize,  although clearly not soon enough. 

 

Works Cited:

Reece, Jane B, and Neil A. Campbell. Campbell Biology. Boston: Benjamin Cummings / Pearson, 2011. Print.

CRISPR Mini | New Territory Unlocked

For over a million years, DNA has centered itself as the building block of life. On one hand, DNA (and the genes DNA makes up) shapes organisms with regard to physical appearance or ways one perceives the world through such senses as vision. However, DNA may also prove problematic, causing sickness/disease either through inherited traits or mutations. For many years, scientists have focused on remedies that indirectly target these harmful mutations. For example, a mutation that causes cancer may be treated through chemotherapy or radiation, where both good and bad cells are killed to stop unchecked cell replication. However, a new area of research, CRISPR, approaches such problems with a new perspective.

The treatment CRISPR arose to answer the question: what if scientists could edit DNA? This technology involves two key components – a guide RNA and a CAS9 protein. Scientists design a guide RNA that locates a specific target area on a strand of DNA. This guide RNA is attached to a CAS9 protein, a molecular scissor that removes the desired DNA nucleotides upon locating them. Thus, this method unlocks the door to edit and replace sequences in DNA and, subsequently, the ways such coding physically manifests itself. Moreover, researchers at Stanford University believe they have further broadened CRISPR’s horizon with their discovery of a way to engineer a smaller and more accessible CRISPR technology.

This study aimed to fix one of CRISPR’s major flaws – it is too large to function in smaller cells, tissues, and organisms. Specifically, the focus of the study was finding a smaller Cas protein that was still effective in mammalian cells. The CRISPR system generally uses a Cas9 protein, which is made of 1000-1500 amino acids. However, researchers experimented with a Cas12f protein which contained only 400-700 amino acids. Here, the new CasMINI only had 529 amino acids. Still, the researchers needed to figure out if this simple protein, which had only existed in Archaea, could be effective in mammals that had more complicated DNA.

To determine whether Cas12f could function in mammals, researchers located mutations in the protein that seemed promising for CRISPR. The goal was for a variant to activate a protein in a cell, turning it green, as this signaled a working variant. After heavy bioengineering, almost all the cells turned green under a microscope. Thus, put together with a guide RNA, CasMINI has been found to work in lab experiments with editing human cells. Indeed, the system was effective throughout the vast majority of tests. While there are still pushes to shrink the mini CRISPR further through a focus on creating a smaller guide RNA, this new technology has already opened the door to a variety of opportunities. I am hopeful that this new system will better the general well-being as a widespread cure to sickness and disease. Though CRISPR, and especially its mini version, are new tools in need of much experimentation, their early findings hint at a future where humans can pave a new path forward in science.

What do you think? Does this small CRISPR technology unlock a new realm of possibility or does it merely shed light on scientists’ lack of control over the world around us?

Clinical Trials to Cure Sickle Cell Disease Using CRISPER Technology

The University of Illinois Chicago participates in clinical trials to cure severe red blood congenital diseases such as sickle cell anemia by safely modifying the DNA of patients’ blood cells. In the CRISPR-Cas9 Gene Editing for Sickle Cell Disease, researchers reported that gene editing modified stem cells’ DNA by deleting the gene BCL11A. This gene is responsible for suppressing fetal hemoglobin production. Then, stem cells start producing fetal hemoglobin so that patients with congenital hemoglobin defects make enough fetal hemoglobin to overcome the effect of the defective hemoglobin that causes their disease.

Sickle cell disease is an inherited defect of the hemoglobin that causes the red blood cells to become crescent-shaped. These cells can lyse and obstruct small blood vessels, depriving the body’s tissues of oxygen.

Sicklecell3

The first two patients to receive the treatment have had successful results and continue to be monitored. Rondelli is on the steering committee for an international clinical trial. The gene manipulation does not use a viral vector as with other gene therapy studies, but this is done with electroporation which is known to have a low risk of off-target gene activation, according to Rondelli. As the strand for the hemoglobin production is very small, being off-target would not allow the treatment to work.

The treatment is created by a small strand of DNA from stem cells that don’t have the gene BCL11A. Researchers do this by editing the strand of DNA by splitting the DNA with a Helicase protein. Then once it is split, it begins to replicate the DNA using small RNA fragments. The researchers then use a specific strand of RNA that does not have a defect. Since they do not have this particular gene, they can produce hemoglobin freely. Now that the cells are producing hemoglobin, they should be able to create enough to stop the blood cells from crescenting. They insert the DNA by electroporation, where the doctors then introduce electronic waves that allow the cell to open. Once the cell is open, the DNA can enter the blood cells.

HemoglobinConformations

This clinical trial is still in its early stages, so it is not used around the world. Though it is promising, it has not been through enough trials. I am not sure if it will get to that stage, do you?

 

Are Genes Inherited from Neanderthals Protecting People Against COVID-19?

Neanderthals, from roughly 40,000 years ago, have had an impact on protecting people, that contain a specific haplotype on chromosome 12, from having severe symptoms due to the Sars-COV-2 virus. Researchers conducted a study that showed a ~22% decrease in severe illness connected to a gene inherited from Neanderthals.   

Neanderthals evolved in western Eurasia -the largest continental area consisting of Europe and Asia- about half a million years ago, living mostly separated from early modern humans in Africa. Neanderthals likely developed certain genes allowing them to fight off infectious diseases during the time of their existence. Due to natural selection, which is when animals with the most favorable traits for survival will survive to reproduce and pass on their genes, these neanderthals were able to evolve and pass on the favorable gene allowing modern humans today to fight off Sars-Cov-2. Through natural selection, the haplotype, on chromosome 12, linked to protection against certain viruses has been passed on. This specific haplotype has helped people during the current pandemic to stay out of the hoHuman male karyotpe high resolution - Chromosome 12spital. 

This study discovered that this specific haplotype on chromosome 12 contains three helpful genes: OAS1, OAS2, and OAS3. These genes encode for a specific enzyme called oligoadenylate synthetase. As we learned in AP Biology, enzymes are created by free ribosomes in the cytosol; the ribosomes manufacture proteins(a chain of amino acids), such as enzymes for cellular reactions. The oligoadenylate chain triggers ribonuclease L. The ribonuclease L, also known as RNase L, is only activated when a viral infection enters the body; it breaks down the viral RNA molecules, leading to autophagy. This enzyme breaks down the viral Sars-Cov-2 RNA and slows/stops the spread of the virus in the body. 

Many people have been trying to find ways to move forward from this pandemic and return to our previous form of normal life. Scientists may be able to use this information about this specific haplotype on chromosome 12 with gene editing technologies, such as CRISPR, to help individuals slow and later stop the spread of COVID-19. Research like this may be one way to be able to return to a normal life-style and keep people out of hospitals from COVID-19. As we continue on in AP Biology this year, I look forward to learning about the idea of genes and gene editing as I will have more knowledge to touch back on this research study. Do you think that this is a possible solution to the COVID-19 pandemic?

 

 

Embryo Gene Editing can Ensure Offspring Do Not Inherit a Deafness Gene!

Denis Rebrikov, A scientist in Russia has done research regarding ways in which he can edit the genome sequence of an embryo in order to prevent the fetus from developing certain gene mutations, specifically in this case a hearing problem or possible complete deafness. His plans are very controversial to some, who believe the possible risks of very harmful mutations to DNA that would be passed onto direct and future offspring, outweigh the possible benefits. However, some people find this scientific possibility to be worth the risk, if it means not passing a potentially very harmful gene down to offspring. If these methods are done correctly, it should alter the genome sequence in the embryo so that future offspring off that embryo will not inherit the negative mutation.

One couple shared their story in detail, in which both parties have a hearing deficiency, the man with partial deafness, and the woman completely deaf. Their biggest hope is to have children who will not inherit hearing issues, because of the apparent challenges they have had to face themselves because of them. They would be the first couple to perform this gene editing on an IVF embryo, so they obviously have some reservations. One of those being publicity, but more importantly the potential risks of using the CRISPR genome editor. They already have a daughter with hearing loss, but they never chose to test her genes for mutations, nor did they get her a cochlear implant to aid her hearing, because of the potential risks of that. When they finally tested her genes, they learned that she had the same common hearing loss mutation called 35delG in both her copies of a gene called GJB2. The parents then tested themselves, realizing they were both 35delG homozygous, meaning their daughter’s mutations were not unique to her, they had been inherited.

If either the mother or father had a normal copy of the GJB2 gene, a fertility clinic could have more easily created embryos by IVF and tested a few cells in each one to select a heterozygote–with normal hearing–to implant. At this stage, Denis Rebrikov informed them that CRISPR genome editing would be their only option. However, the process presents possibly deal breaking risks, such as mosaicism, in which a gene edit might fail to fix the deafness mutation, which could create other possible dangerous mutations like genetic disorders or cancer. The couple has not decided to go through with the editing just yet, but it is something they are open to in the future as more possible new research or test subjects become available.

Explaining the CRISPR Method: “The CRISPR-Cas9 system works similarly in the lab. Researchers create a small piece of RNA with a short “guide” sequence that attaches (binds) to a specific target sequence of DNA in a genome. The RNA also binds to the Cas9 enzyme. The modified RNA is used to recognize the DNA sequence, and the Cas9 enzyme cuts the DNA at the targeted location… Once the DNA is cut, researchers use the cell’s own DNA repair machinery to add or delete pieces of genetic material, or to make changes to the DNA by replacing an existing segment with a customized DNA sequence.” -US National Library of Medicine Genetics Home Reference

l

Woman with a hearing aid 

If you had the opportunity to alter something in the gene’s of your baby’s embryo, would you? Under what circumstances would you consider this, and what risks might stop you from deciding to do it? Comment down below.

 

 

The First “in vivo” CRISPR-Cas9 Gene Editing

At the start of the new year all of the scientists working in different fields being to create a schedule of perceived accomplishments that will occur in regard to their specific field of study. In the term of the people working with CRISPR, they speculate that the first in body injection in order to conduct real time genome editing will occur in 2020. Prior to this year, CRISPR has been used to edit and alter the DNA of red blood cells outside of the body, but the scientists working with this new form of biological technology belief that this year it will be used the way it is intended to be used.

CRISPR was originally found in 2012 inside bacteria in order to help stop viruses from infecting them. Scientists saw the possible benefit of this in humans and through years of research discovered that CRISPR can help place another enzyme Cas9, which snips out parts of DNA, in a correct spot on the genome to alter and edit a person’s DNA code. While over time some scientists have wondered about the ethics of this new discovery, most are excited by all of the possibilities that CRISPR has on curing diseases.

Currently, the reason that CRISPR-Cas9 has become a talking point recently was due to the fact and difference between gene editing “in vivo” versus gene editing “ex vivo”, meaning within the body or outside the body. Scientists working with CRISPR have been able to understand how editing “ex vivo” works. They are able to see the genes they want to edit and watch the process occur. This is also easier for the scientists because if the editing messes up, they do not need to reinsert the altered cell back into the body. On the other hand, “in vivo” gene editing is much more efficient and can be completed through a simple injection, but may cause dangerous consequences, such as cancer, if a mistake is made in both the scientists’ coding of the CRISPR or in the process done by the CRISPR itself.

Now that scientists are starting to attempt to move from “ex vivo” editing to “in vivo” editing, all of these questions and issues are being brought up. With lots of labs around the country working on moving into “in vivo” editing and an FDA approval for the procedure, the first CRISPR-Cas9 “in vivo” gene editing is bound to happen soon. Hopefully, this new biological technology does its job properly and gives hope to those who have various currently seemingly incurable disease. If successful, CRISPR could revolutionize medicine itself to make it more efficient and effective. Feel free to comment about how you think CRISPR will do in the first “in vivo” test and how it could effect life later on.

CRISPRi Antibiotics: Will Pathogens Cease to Exist?

Recently, a researcher at the University of Wisconsin-Madison and his collaborators at the University of California, San Francisco have discovered a way to repurpose CRISPR, a gene-editing tool, to develop new antibiotics.

What does this mean?

It is known that many disease-causing pathogens are resistant to current antibiotics. This new technique, Mobile-CRISPRi, is helping to change that. Mobile-CRISPRi allows scientists to “screen for antibiotic function in a wide range of pathogenic bacteria.” Scientists used of bacterial sex to transfer Mobile-CRISPRi from laboratory strains into any diverse bacteria. This easily transferred technique will now allow scientists to to study any bacteria that cause disease or help one’s health.

To break it down, Mobile-CRISPRi “reduces the production of protein, from targeted genes, allowing researchers to identify how antibiotics inhibit the growth of pathogens.” This will allow researchers to more thoroughly understand different bacterias’ resistance to current antibiotics.

Unlike CRISPR, which can split DNA into two halves, CRISPRi is a defanged form that is unable to cut DNA. Instead, it stays on top of the DNA, blocking other proteins from being able to turn on a specific gene.

Genes (DNA)

Why is this important?

It has been proved that a decrease in the amount of protein targeted by an antibiotic will make bacteria become more sensitive to lower amounts of that same antibiotic. This evidence association between “gene and drug” has allowed scientists to “screen thousands of genes at a time,” as they try to gain more knowledge about the mechanisms of how antibiotics work in organisms to improve those on the market now.

In order to study how antibiotics directly work in pathogens, researchers needed to make this CRISPRi mobile, or easily transferred onto different bacterias. There were two tests done to test CRISPRi’s mobility using conjugation. One involving a transfer of CRISPRi to the pathogens Pseudomonas, Salmonella, Staphylococcus, Listeria, and others, and another involving the bacteria that grows on cheese.

Science Daily describes the “landscapes of microbes cheese creates as it ages.” A bacteria called “Vibrio casei” was found on a French cheese in a lab back in 2010. With bacteria that have been taken out of their environment, it is hard to manipulate and study said pathogen’s genes, like “Vibrio casei.”

However, the mobile-CRISPRi was able to easily transfer onto the strain of  Vibrio casei that was discovered back in 2010. This has given scientists a new way of understanding how bacteria colonizes and ages cheese.

Peters, the head researcher of mobile-CRISPRi is generous enough to offer this system to other scientists who would like to study other germs. With this technology being available to others, scientists could potentially see first-hand how antibiotics work in pathogens, and use that information to improve those that currently exist, hopefully getting rid of these bacterias like pseudomonas, or listeria before they kill the human they are inside of.

CRISPR Cas9, too good to be true?

After its peak in popularity following its reveal as a possible “genetic modifier” in 2013, the CRISPR Cas-9 enzyme system has been the center of debate within the biology community. Thought to be the solution to all genetic and hereditary diseases by simply “cutting out” the fault gene, new research and studies have shown that a majority of people (65 to 79 percent) have antibodies that would fight cas-9 proteins.

“The study analyzed blood for antibodies to two bacteria from which Cas9 is derived: Streptococcus pyogenes and Staphylococcus aureus. The researchers’ concern stemmed from the fact that these bacteria frequently cause infections in humans, and so antibodies to them may be in our blood” states bigthink.com

While the overall effects are unclear, the study concludes that the result would be “significant toxicity” and an unsafe use of the gene editing tool.

What do you think? Is the current risk of using Cas-9 worth the reward?

Click here, here, and here for more information.

Cas9, photo by J LEVIN W

 

CRISPR, A Cure to Heart Disease?

Photo Source page: Flickr.com

     While CRISPR‘s full potential in the department of gene editing is still being researched, scientists have just successfully discovered CRISPR’s ability to correct a defective gene that causes a certain type of heart disease. Though scientists are unclear as to the type of gene corrected in order to cause this change, this discovery was made for the first time in the United States, by an experiment done on live human embryos. However the new information yielded from this experiment is extremely beneficial as it shows CRISPR’s potential in correcting genetic errors that cause disease, as well as in human embryos meant for pregnancy.

Another reason for which this study particularly stands out in its importance, is because it is much different from the other developments scientists have made in CRISPR’s abilities. Studies have been conducted worldwide using CRISPR to edit of somatic cell’s gnomes, however, this only affects individual people. This study (also done by researchers in China), has been done by editing germ line cells, which result in changes that are passed down through every following generation.

However since the changes made to cells do affect all generations that follow, scientists are unsure of the exact effects of this new technique. Although it seems that this technology will be very beneficial in stopping harmful genetic diseases, it can also be used for changing DNA to genetically determine the eye colors, height or even mental and physical abilities and intelligence. This new phenomenon is own as “designer babies”, and for many reasons, this is not something that the United States is trying to use CRISPR’s abilities for. For this reason, United States has recently created more severe guidelines regarding gene editing technology, as well as enforcing CRISPR’s use on embryos only for prevention of harmful genetic diseases, when other treatments were not successful – as a last resort – formed by the National Academies of Sciences, Engineering and Medicine.

In the study done, scientists edited out a mutant copy of MYBPC3, using CRISPR. MYBPC3 is a gene that encodes a protein that creates well maintained and structured heart muscles. Hypertrophic cardiomyopathy, known as HCM, are caused by mutations in that gene, and cause spontaneous cardiac arrest. This occurs in even the youngest and healthiest of athletes, affecting 1 in 500 humans.

In this study, the mother was carrying the normal version of a gene, while the father had the mutant gene. Using CRISPR, the scientists were able fix the mutant version, by cutting and replacing the DNA. Directly after they placed the fertilized egg in a petri dish, while introducing the genome editing parts at the same time. The results of this process proved to be very effective, as 75% of the embryos showed no mutant genome. Without the use of CRISPR when egg fertilization occurred, the chances of mutation would have been present in 50%!

From these results the researchers came to the conclusion that they have realized the potential for mosaicism. Mosaicism is when only some of the cells are edited and the rest are not affected, which results in some normal cells, as well as some mutant cells. The scientists have also gathered the effects of off-targets. Off targets are the CRISPR edited genes that appear to look like mutant genes, but are actually not. Within this study, one egg fertilized from 58 showed mosaicism, and there was no detection of effects from off-targets. Theseare very impressive results, due to the fact that both of these possible situations can cause limitations in effectiveness and safety.

Though researchers need to do over this experiment many times in order to soliditfy the effectiveness of this study for the future, if they want to use this on eggs intended for pregnancy, as the eggs fertilized in the study were not meant for pregnancy… However, the results have yielded nothing but good news for the future of CRISPR technology (besides, the risk in advancements in “designer babies”, which couldchange the future of conceiving, forever…). This article was extremely interesting for me to read, as I am very interested in studying Biology in the future, or even pursuing a track to medicine. Perhaps, I may get the chance to even experiment with CRISPR at some time in my life, as it becomes a growing presence throughout the science world!

Primary Source Article: U.S. researchershave used gene editing to combat heart disease in human embryos

HIV Adapts to CRISPR-Cas9 Treatment

There has been an abundance of research using CRISPR/Cas9 gene editing to search for a cure for HIV. The HIV virus enters immune cells and uses the host cell’s method of replication to replicate the viral genome. With CRISPR/Cas9, specific mutations can be introduced in order to make it more challenging for the HIV virus to enter Helper T-Cells. Guided by specific strands of RNA, the Cas9 enzyme can cut a particular piece of the viral genome out, rendering it useless.

When a team of researchers at McGill University attempted to use the CRISPR method to disable the HIV viral genome, they found a major roadblock. Two weeks after the CRISPR/Cas9 treatment, the host cells appeared to be creating copies of the virus. This may be attributed to an error in the enzymes that copy the viral DNA, causing a change in the genome, and a mutation that allows it to evade the CRISPR treatment. However, the McGill researchers believe that this mutation was a result of the CRISPR treatment itself.

After DNA is cut by the Cas9 enzyme, the host cell usually attempts to repair the damage. Occasionally, this results in the addition or deletion of a few nitrogenous bases. While these changes usually result in the inactivation of the cut gene, sometimes they don’t. The active cut DNA is no longer recognized by the machinery used to prevent HIV infection of the cell, and the mutated viral genome is resistant to the usual methods of disablement.

More researchers at the University of Amsterdam had similar results in their research. While it is not that surprising that HIV can overcome the CRISPR/Cas9 gene editing at some point, the leader of the research (Atze Das) said “What is surprising is the speed- how fast it goes”.

If CRISPR was used at the same time as HIV-attacking drugs (inhibitors of protease, reverse transcriptase, and integrase), perhaps the mutations would be less  detrimental. This roadblock does not mean that a CRISPR cure for HIV is impossible, but it does make it far more challenging to overcome.

The Grey Area of Human Gene Editing

The process of Human Gene Editing developed with the goal to prevent future generations from suffering from genetic diseases present in past generations, like our own. Human gene editing, provided it is done only to the correct disease, alters the DNA in embryos, eggs, and sperm to the when reproduction occurs, the gene for the disease or disability is not inherited. However, two weeks ago the National Academies of Sciences and Medicine issued a report stating that human gene editing is being used to enhance people’s health or abilities. This is considered unethical according to organizers of a Global Summit on human gene editing.

Human gene editing has been given a “yellow light” because the process is not yet approved to be done on people. There are high hopes that diseases caused by only 1 genetic mutation such cystic fibrosis and Huntington’s disease will be eliminated due to this process. Unfortunately diseases that are caused by more than one genetic mutation, such as autism or schizophrenia, are not curable by this process.

National Cancer Institute

Gene Editing on humans is such a controversial topic right now: is it ethical to change genes? should the practice be used to change physical appearances? Ultimately, if Human Gene Editing is approves, who decides when it becomes too much, or unethical. This grey area is presented to be somewhere between when it is appropriate to help aid the life of a human, ridding them of a disease, and when enhancements are made.

 

Powered by WordPress & Theme by Anders Norén

Skip to toolbar