Engineer ourself and nature: CRISPR
Giulia Sironi
Genetic editing is not just rewriting DNA.
It’s quietly rewriting our relationship with nature.
For the first time, we can directly intervene in the code of life.
Modify, correct, improve.
It’s a tremendous possibility.
But also a new balance we must imagine: between control and respect, between what we can do and what we want to become.
Because every modification carries an invisible responsibility.
It calls for awareness.
The truth is, we still don’t know enough.
About ourselves.
About the world.
About the chain reaction that even the smallest change can trigger in an entire ecosystem.
Over the past decade, crispr–cas gene-editing technology has evolved from an academic curiosity into a powerful tool with the potential to revolutionize medicine. since the discovery of its extraordinary capabilities, crispr has traveled a surprising path, becoming the symbol of a new era in biotechnology. but despite the promises and some notable successes, its widespread clinical adoption remains an open frontier.

From origins to molecular precision
The turning point came in 2012, when a team led by emmanuelle charpentier and jennifer doudna published a groundbreaking study demonstrating that the crispr–cas9 system could cut specific dna sequences in a targeted way. since then, the field has rapidly evolved. The introduction of variants such as cas12 and cas13 has expanded the system’s capabilities, from dna modification to rna manipulation, opening up new application scenarios.
But what exactly is crispr?
The acronym stands for clustered regularly interspaced short palindromic repeats — sequences that function as a bacterial immune system, defending against viruses. scientists have learned to harness this natural mechanism to edit the genome of complex organisms with remarkable precision.
The system uses a guide rna (grna), designed to recognize a specific dna target. this guide forms a complex with an enzyme — usually cas9 — which acts as a “molecular scissor,” cutting the dna at a precise point. the cell’s natural repair mechanisms are then used to make changes: deletions, insertions, or even precise corrections.
In recent years, new variants such as cas12 have brought improvements in specificity, reducing the risk of unwanted cuts and expanding the range of genetic targets. cas13, on the other hand, enabled direct editing of rna for the first time, offering the possibility to temporarily influence gene expression without permanently altering the dna.
A technological leap: base editing and prime editing
If Crispr–cas9 opened a new era, more recent developments are set to refine this revolution even further. Harvard researcher David Liu introduced two complementary techniques: base editing and prime editing. base editing allows for the correction of single “letters” in the genetic code without cutting the dna double helix, minimizing the risk of errors. prime editing, even more sophisticated, enables precise modifications without causing breaks in the dna, making therapeutic interventions potentially safer.
These tools have already shown promising results in preclinical models and are paving the way for highly precise genetic therapies for rare diseases, inherited disorders, and, in the future, even complex conditions like alzheimer’s or diabetes.

A clinical milestone: Casgevy and the future of gene therapy
A major milestone arrived in 2023, when the crispr-based treatment casgevy (exagamglogene autotemcel) received regulatory approval — first in the uk, then in the us — for use in treating beta-thalassemia and sickle cell anemia. This approval is not only a scientific victory but a clear signal that the transition from lab to clinic is possible.
Casgevy works by modifying the patient’s hematopoietic stem cells. once extracted, these cells are edited ex vivo to reactivate the production of fetal hemoglobin, a type of hemoglobin that compensates for the genetic defect at the root of these diseases. when reinfused into the patient, the edited cells correct the issue at its core. it’s a step that could radically change how we approach genetic disorders.
But that’s not all. Innovative startups like tune therapeutics are developing epigenetic technologies, such as tune-401, which aim to silence the viral dna responsible for hepatitis b without cutting it. this strategy avoids the challenges of permanent genome editing and reduces potential side effects.
The delivery dilemma
Despite these breakthroughs, one of the most complex challenges remains the delivery of crispr components to target cells. viral vectors, such as adeno-associated viruses (aavs), are the most commonly used method but come with serious limitations: limited payload capacity, high production costs, and possible immune responses.
alternatives such as lipid nanoparticles (lnps), already used successfully in mrna vaccines, are under study. they offer greater safety and flexibility, but their efficiency remains suboptimal in many cell types and tissues. the challenge is to develop systems capable of crossing the blood-brain barrier, penetrating hard-to-reach organs like the brain, and doing so with almost surgical precision.
Scalability, safety, and ethics: a revolution waiting for an infrastructure
beyond delivery, scalability is another significant hurdle. consistently producing crispr components and associated vectors on an industrial scale requires sophisticated facilities, rigorous quality controls, and high costs that currently limit access to treatments to a select few. in this context, democratizing genetic care remains an open issue.
safety is another major concern. off-target effects — unintended genetic changes — can have serious consequences, including the potential onset of cancer. furthermore, since cas proteins are of bacterial origin, they can trigger immune reactions that undermine the treatment’s effectiveness.
these risks raise deep ethical questions. where is the line between healing and enhancement? is it acceptable to modify the germline — the dna passed to future generations? how do we prevent a revolutionary technology from becoming the privilege of a few?
The path ahead, where science and market meet
The biotech industry’s interest in crispr has exploded. in 2024, the total value of the gene-editing market surpassed $10 billion, with exponential growth projected. companies like crispr therapeutics, editas medicine, and intellia therapeutics are at the forefront, developing treatments for rare genetic diseases — but also for more common conditions like high cholesterol and certain cancers.
The future of crispr is both exciting and complex. in the coming years, we may see personalized therapies developed in a matter of weeks, editing of the gut microbiome to treat metabolic disorders, or even genetic interventions at the embryonic stage to correct diseases before birth.
But to fully realize crispr’s potential, we must address a series of interdisciplinary challenges — scientific, engineering, ethical, and social. integrating emerging technologies — from artificial intelligence to bioinformatics — will be crucial to enhance precision and minimize risks.
Like every major innovation, crispr confronts us with fundamental questions about our place in the world and the need to accept how little we still know about it.

Giulia Sironi is a writer, TEDx speaker, and from Milan, Lombardy, Italy
Mahabahu.com is an Online Magazine with collection of premium Assamese and English articles and posts with cultural base and modern thinking. You can send your articles to editor@mahabahu.com / editor@mahabahoo.com(For Assamese article, Unicode font is necessary) Images from different sources.