Search Immortality Topics:

Page 11234..1020..»


Nanoscope Therapeutics: Gene Therapy Improves Visual Acuity in Patients with Retinitis Pigmentosa – 2 Minute Medicine

Posted: May 6, 2024 at 2:47 am

The Latest

A recent two-year phase 2b, randomized, double-masked, sham-controlled multicenter clinical trial by Boyer et al. and Nanoscope Therapeutics investigated mutation-agnostic gene therapy for the treatment of permanent and severe vision loss from of retinitis pigmentosa. The results build on an earlier trial that found 89% of patients injected with the gene therapy to experience an improvement of luminance levels across two visual tests compared to control group patients. This newest trial, named RESTORE, demonstrated significant improvement in best-corrected visual acuity after 52 weeks compared to the control arm. The results showed that gene therapy was well tolerated, with no treatment-related serious or severe adverse events reported.

Physicians Perspective

Retinitis Pigmentosa encompasses a group of rare genetic eye disorders in which the retinas photoreceptors degrade over time leading to profound visual field and vision loss in advanced stages. Retinitis pigmentosa affects 1 in every 400 people in the United States and approximately 1 in 5000 worldwide, making it the most common inherited disease of the retina. There are currently no cures available for retinitis pigmentosa. Current gene therapies aim to treat patients with specific gene mutations and are limited in advanced disease with degenerated outer retinal cells. Nanoscopes optogenetic monotherapy targets the intact inner retinal neurons to restore vison loss. This approach has the advantage of restoring vision even in advanced retinitis pigmentosa, regardless of causative gene mutation. Furthermore, the therapy is administered via a single intravitreal injection without any need for external devices.

Molecular Targets

Nanoscope Therapeutics has developed a gene therapy called MCO-010 that uses light sensitive molecules to treat retinal disease. MCO-010 is an injection that transforms bipolar cells that normally do not transmit light (are not sensitizing) to become light sensitizing. The gene therapy works by transfecting the cell layers above the damaged cone layers, such as the bipolar and ganglion cells, into viable light producing cells. MCO-010 is activated by ambient light across the visual spectrum.

Company History

Nanoscope therapeutics is a Texas based late-stage clinical biotechnology company developing gene therapies for inherited retinal diseases and age-related macular degeneration. MCO-010 is the companys lead asset and has recently received FDA fast-track designations. Additionally, the company has recently completed a phase 2 trial of MCO-010 in Stargardt disease.

Further reading: https://www.fiercebiotech.com/biotech/nanoscope-eyes-market-after-gene-therapy-improves-vision-patients-retinal-disease

2024 2 Minute Medicine, Inc. All rights reserved. No works may be reproduced without expressed written consent from 2 Minute Medicine, Inc. Inquire about licensing here. No article should be construed as medical advice and is not intended as such by the authors or by 2 Minute Medicine, Inc.

Continued here:
Nanoscope Therapeutics: Gene Therapy Improves Visual Acuity in Patients with Retinitis Pigmentosa - 2 Minute Medicine

Recommendation and review posted by G. Smith

College of Engineering Launches New Collaboratory for Biomedical and Bioengineering Innovation – UConn Today – University of Connecticut

Posted: May 6, 2024 at 2:47 am

A new initiative in the College of Engineering will serve as the nexus for bio-based technology at UConn.

The Collaboratory for Biomedical and Bioengineering Innovation fosters a vibrant and unified environment where biomedical and bioengineering researchers work together to invent, develop, and adapt existing biotechnologies to solve new problems in the biological sciences.

There seems to be an artificial divide between researchers who focus on biomedical studies and those working on other biological problems, says Leslie Shor, associate dean for research and graduate education and co-director of Collaboratory for Biomedical and Bioengineering Innovation. This is especially strange for engineers, because we are often leading the technical aspects of the work, and an enabling technology such as a novel sensor or new imaging technology works the same regardless of the biological application.

The Collaboratory, however, aims to help researchers establish new interdisciplinary collaborations outside their existing research networks.

By promulgating emerging technologies across fields, we enhance the value of the emerging technology and simultaneously unlock new areas of inquiry and accelerate new discoveries, Shor explains.

Bio-based technology, or biotechnology innovation refers to the development and advancement of technologies that are based on biological systems or use biological materials. This can include a wide range of innovations such as biomedical devices (prosthetics, medical imaging equipment, drug delivery systems); bio-systems (biofuels production, bioremediation of pollutants, agricultural biotechnology); and bio-computation (bioinformatics for analyzing genetic data, computational modeling of biological systems, or machine learning algorithms for drug discovery).

Members of the Collaboratory are nationally and internationally-renowned faculty.

Thanh Nguyen, associate professor of mechanical engineering and biomedical engineering, works at the interface of biomedicine, materials and nano/micro technology. Hes already collaborating with researchers on campus and UConn Health for vaccine, drug, tissue-engineering and biomaterials research, but expects the Collaboratory for Biomedical and Bioengineering Innovation will help strengthen those relationships and allow him to explore more research opportunities.

UConn is already a collaborative and terrific environment for interdisciplinary research. But this initiative makes biomedical and engineering research from different groups much more visible to all researchers at UConn. Nguyen says. The Collaboratory also could eventually lead to more impactful studies and grant funding.

Like Nguyen, Sabato Santaniello, associate professor of biomedical engineering, is interested in potential collaborations with UConn Health and other medical centers in the region. His work in neuromodulation of the cerebellum is primarily targeted to clinical neuroscienceproviding new ways of probing the diseased brain and improving treatments of patients affected by movement disorders.

My work has potential to translate into new, patentable products down the road, but now, my program can benefit the initiative by intercepting the needs of clinicians, especially neurologists and neurosurgeons, he says.

Santaniello describes the Collaboratory as a unique platform that will regionally advertise the many cutting-edge biomedical technologies that UConn faculty develop and better intercept the needs that come from the healthcare industry and the clinical research.

It will benefit greatly those PIs at UConn who are looking for new, exciting applications for the tools that are developed in their labs, he says.

The group aims to promote bio-based technologies through collaborative research; boost economic growth in Connecticut by creating new bio-based products and businesses; train students for biotech careers by involving them in research and innovation; and establish UConn as a global leader in bio-based technology innovation.

Our goals are to drive research, investment, and possibilities in Connecticut, explains Guoan Zheng, associate professor of biomedical engineering and co-director of the Collaboratory for Biomedical and Bioengineering Innovation. By advancing technology, we believe we can make a significant impact on scientific discovery and its applications driving socially impactful research and benefiting Connecticuts economy and workforce.

Shor, whos also Centennial Professor of Chemical and Biomolecular Engineering, leads the Engineered Microhabitats Research Group at UConn, where she mentors an interdisciplinary team focusing on biotechnology for sustainability. My lab simply adapted established microfluidics or lab-on-a-chip technologies to a completely different field of biology: soil microbes living near plant roots. This approach directly led to new understanding about soil moisture regulation by bacteria and fungi and a new appreciation for how soil protists can be used to promote more sustainable food production. I want to see the same interchange of approaches advance all types of biological sciences to advance a healthy and sustainable future, she said.

The Collaboratory is seeking student, faculty, and corporate partners. For more information, contact the UConn Collaboratory.

The Collaboratory for Biomedical and Bioengineering Innovation celebrated its launch April 2 with a networking symposium and poster session. Faculty from several engineering disciplines attended to learn about the interdisciplinary relationships related to biomedical and bioengineering research and technology innovation. Photos of the event are below and in this UConn College of Engineering Flickr album. (Chris LaRosa/UConn)

See more here:
College of Engineering Launches New Collaboratory for Biomedical and Bioengineering Innovation - UConn Today - University of Connecticut

Recommendation and review posted by G. Smith

"The best decision I ever made:" Patel earns degree in biological engineering – University of Missouri College of Engineering

Posted: May 6, 2024 at 2:47 am

May 03, 2024

Zara Patel didnt think she would find herself at Mizzou, but after four years on campus she says attending school here was the best decision shed ever made.

Patel will graduate with a degree in biological engineering, which she chose because of her passion for creating technology that has a positive environmental impact. Outside of the classroom, shes been involved in multiple student organizations with focuses on both academics and college traditions.

After graduation, she will begin her career as a water/wastewater designer at Stantec in Indianapolis.

Read on for a Q&A about her time at Mizzou.

Why did you choose Mizzou?

I was born and raised here in Columbia, Missouri. Both my parents went to Mizzou to get their undergraduate degrees and then stayed once they graduated. I never really thought that I was going to go to Mizzou. I always assumed that I would leave the state for college, but once the pandemic began, it was more difficult to go to a school that was out of state and I decided to go to Mizzou. It was one of the best decisions Ive evermade.

What made you interested in your major?

I originally started at Mizzou as a biological sciences major and then switched to biological engineering with an emphasis in bioenvironmental engineering. I always knew I wanted to major in a STEM field and that I wanted to make a difference. I switched majors because I wanted to have a greater connection with creating technology that has a positive impact on the environment, specifically focusing on biological integrations.

How did you get involved at Mizzou?

I am involved in Alpha Omega Epsilon, an engineering and STEM sorority. I am also involved in the Society of Sales Engineers and Engineers Club. Getting to know all the Engineers Week royalty candidates personally, as I was on the royalty committee for the Engineers Club, allowed for me to fully get immersed in the skits. That was my favorite Mizzou Engineering memory.

Whats next for you after graduation?

I have accepted a position at Stantec as a water/wastewater designer in Indianapolis.

What would you tell someone whos interested in coming to Mizzou?

Mizzou is about community and the environment. Whether you come to the school knowing someone or as a total stranger, you will always make friends. Every university has an environment, but Mizzous environment is differentyou can find any group that you want. The first time you fully emerge yourself in the environment, whether it be in classes, student org meetings or at a game, you will know that you made the right choice.

Read this article:
"The best decision I ever made:" Patel earns degree in biological engineering - University of Missouri College of Engineering

Recommendation and review posted by G. Smith

Investigation of inherited noncoding genetic variation impacting the pharmacogenomics of childhood acute … – Nature.com

Posted: May 6, 2024 at 2:47 am

Identification of noncoding regulatory variants impacting the pharmacogenomics of ALL treatment

Single-nucleotide variants (SNVs) impacting diverse pharmacological traits in ALL were identified for functional interrogation. We chose SNVs associated with relapse or persistence of MRD after induction chemotherapy in childhood ALL patients to investigate the role of inherited noncoding regulatory variants impacting clinical phenotypes (i.e., treatment outcome). These SNVs were identified from published GWAS of ALL patients enrolled in St. Jude Childrens Research Hospital and the Childrens Oncology Group clinical protocols3,4,5 (see Methods for variant selection criteria). Variant selection also included prioritization for treatment outcome SNVs associated with drug resistance phenotypes in primary ALL cells to enrich for variation impacting ALL cell biology (see Methods for variant selection criteria). These treatment outcome-associated variants, as well as all variants in high LD (r2>0.8) with the sentinel GWAS variants, were further evaluated (Fig.1a, b).

a SNVs of interest from GWAS were pursued based on association with ex vivo chemotherapeutic drug resistance in primary ALL cells from patients and/or treatment outcome. Dex dexamethasone, Pred prednisolone, VCR vincristine, 6MP 6-mercaptopurine, 6TG 6-thioguanine, LASP L-asparaginase. b GWAS SNVs were combined with ALL disease susceptibly control GWAS SNVs and SNVs in high LD (R2>0.8) and c mapped to accessible chromatin sites in ALL cell lines, ALL PDXs, and primary ALL cells from patients. Of the 1696 SNVs mapped to accessible chromatin sites, 35 are control SNVs. Source data are provided in the Source Data file.

We also identified variants directly associated with ex vivo chemotherapeutic drug resistance in primary ALL cells from patients by performing GWAS analyses using SNV genotype information and ex vivo drug resistance assay results for six antileukemic agents (prednisolone, dexamethasone, vincristine, L-asparaginase, 6-mercaptopurine [6MP] and 6-thioguanine [6TG]) in primary ALL cells from 312344 patients (not all patients were tested for all drugs) enrolled in the Total Therapy XVI clinical protocol at St. Jude Childrens Research Hospital (see Methods). We further prioritized functional ex vivo drug resistance SNVs by determining if they were eQTLs in primary ALL cells or related cell types (i.e., whole blood and EBV-transformed lymphocytes) from the Genotype-Tissue Expression (GTEx) consortium37 (see Methods for variant selection criteria). Ex vivo drug resistance variants that were also identified as eQTLs, as well as variants in high LD (r2>0.8) with these sentinel GWAS variants, were further evaluated (Fig.1a, b).

GWAS have also been performed for childhood ALL disease susceptibility and identified several GWAS loci harboring variants with genome-wide significance44,45,46,47,48,49,50. Several follow-up studies of these GWAS loci have identified candidate causal noncoding variants and mechanisms involving gene regulatory disruptions51,52,53. As a result, we used ALL disease susceptibility variants (n=11), as well as variants in high LD (r2>0.8) with them, for further analysis as positive controls in our study (Fig.1a, b).

Because most of these variants map to noncoding portions of the human genome, these data point to disruptions in gene regulation as the underlying mechanism of how these variants impact ALL cell biology. We therefore utilized assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq)54 chromatin accessibility data in 161 ALL cell models, comprised of primary ALL cells (cryopreserved, n=2455; fresh, n=12056), ALL cell lines (n=14) and ALL patient-derived xenografts (PDXs, n=3), to uncover which variants map to putative CREs in ALL cells57 (i.e., regulatory variants; Fig.1c). Although we detected variation in ATAC-seq TSS enrichment scores and peak counts that is to be expected from such a large, mixed cohort of ALL cell models, the peaks called were largely reproducible (found in >3 samples) within each group (Supplementary Fig.1ac). ATAC-seq data from primary ALL cells, ALL cell lines, and PDXs were combined and identified 1696 regulatory variants at accessible chromatin sites in ALL cells for functional investigation (Fig.1c and Supplementary Data1).

To examine the functional effects of these 1696 regulatory variants on transcriptional output in a high-throughput manner we utilized a barcode-based MPRA platform29,32 to measure differences in allele-specific transcriptional output (Fig.2a). Oligonucleotides containing 175-bp of genomic sequence centered on each reference (ref) or alternative (alt) variant allele, a restriction site, and a unique 10-bp barcode sequence were cloned into plasmids. An open reading frame containing a minimal promoter driving GFP was then inserted at the restriction site between the alleles of interest and their unique barcodes (Fig.2a). We utilized 28 unique 3UTR DNA barcodes per variant allele (56 barcodes per regulatory variant), and variants near bidirectional promoters (47 total variants) were tested using both sequence orientations. In total, 97,608 variant-harboring oligonucleotides were evaluated for allele-specific differences in gene regulatory activity (Fig.2a).

a Diagram describing design of MPRA (also see Methods). bd Significant MPRA hits were identified by BenjaminiHochberg FDR corrected two-tailed Students T tests. b Distribution of significant changes in allele-specific transcriptional activity across all SNVs. c Number of MPRA SNVs showing significant (Adj. p<0.05) changes in allele-specific transcriptional activity in each ALL cell line. d Pairwise linear correlation between changes in allele-specific transcriptional activity for all significant (Adj. p<0.05) changes across all cell lines. R2 correlation and p value are provided. All source data and statistical parameters are provided in the Source Data file.

Following transfection into 7 different B-cell precursor ALL (B-ALL; 697, BALL1, Nalm6, REH, RS411, SEM, SUPB15) and 3 T-cell ALL (T-ALL; CEM, Jurkat, P12-Ichikawa) human cell lines (n=4 transfections per cell line; 40 total), the transcriptional activity of each allele variant was measured by high-throughput sequencing to determine the barcode representation in reporter mRNA and compared to DNA counts obtained from high-throughput sequencing of the MPRA plasmid pool (Fig.2a). In the 10 cell lines MPRA detected 4633 instances of significant differential activity between alleles across 91% (1538/1696) of regulatory variants tested (Fig.2b, c, Supplementary Data2). The 10 ALL cell lines showed substantial differences in the total number of regulatory variants harboring significant allele-specific activity, which we suspect largely stems from differences in transfection efficiency (Fig.2c). Importantly, when comparing changes in allele-specific MPRA activity for each regulatory variant we found that significant changes in activity (adj. p<0.05) were highly correlated between ALL cell lines, with 87% concordance in allelic-specific activity, suggesting that significant MPRA hits were likely to be robust and reproducible between cell lines (Fig.2d). Allele-specific MPRA activities were also correlated using all pairwise cell line comparisons for each regulatory variant, irrespective of significance (Supplementary Fig.2a). Importantly, 31 of the 35 positive control variants (i.e., ALL disease susceptibility-associated variants and variants in high LD) showed significant allelic effects in at least 1 cell line, and 10 showed significant and concordant allelic effects in at least three ALL cell lines, including two variants (rs3824662 at GATA3 locus and rs75777619 at 8q24.21) directly associated with ALL susceptibility44,49,52 (Supplementary Data2). The risk A allele at rs3824662 was associated with higher GATA3 expression and chromatin accessibility and demonstrated significantly higher allele-specific activity in our MPRA44,52, thereby demonstrating that the MPRA could detect allelic effects previously identified by others.

To further validate MPRA hits in an ex vivo model, we performed MPRA using two B-ALL PDX samples that were freshly harvested from mice. These samples detected 26 and 67 significant gene regulatory variants, respectively, and showed significant correlation with the cell line MPRA data (Supplementary Fig.2b, c, Supplementary Data3). We attribute the detection of relatively lower numbers of variants in PDXs to technical effects stemming from poor transfection efficiency and limited cell survival ex vivo. Overall, our data suggest that the cohort of SNVs tested contained functional regulatory variants with the potential to impact gene regulation.

To further focus on regulatory variants most likely to broadly impact gene regulation in ALL cells, we prioritized 556 variants with significant (adj. p<0.05) and concordant allele-specific activities in at least three ALL cell lines (i.e., functional regulatory variants; Fig.3ad, Supplementary Data4). Most of these functional regulatory variants (318/556) mapped to accessible chromatin found only in primary ALL cell samples, underscoring the importance of incorporating chromatin architecture from primary ALL cells, and 54 functional regulatory variants mapped to transcription factor footprints in primary ALL cells (Supplementary Fig.3). Additionally, we used Genomic Regions Enrichment of Annotations Tool (GREAT) to associate these SNVs with their nearby genes and search for enrichment in gene ontology biological processes pathways58. Although GREAT identified gene associations for nearly all SNVs, we found no significant pathway associations (Supplementary Data4 and 5). Because further functional investigation of variants in primary ALL cells or PDXs ex vivo is largely intractable, we focused on 210 functional regulatory variants that were detected in open chromatin in one of the 14 ALL cell lines that we had generated ATAC-seq data (Fig.3d). Most of these variants (159/210; 76%) were also found in accessible chromatin in PDXs and/or in primary ALL cells from patients (Fig.3d).

a 556 of the 1696 SNVs assayed are functional regulatory variants with reproducible (FDR<0.05 in >2 cell lines) and concordant (same directionality in >2 cell lines) changes in allele-specific activity. b Frequency distribution plot showing the number of cell samples showing concordant and significant MPRA activity of variants. c Plot showing the distribution of log2-adjusted activity between alternative (Alt) and reference (Ref) alleles across 556 functional regulatory variants. 210 SNVs (in blue) mapped to accessible chromatin sites in ALL cell lines and 346 SNVs (in black) mapped only to accessible chromatin sites identified in primary ALL cells and/or PDXs. d Upset plot shows how many functional regulatory variants map to open chromatin in diverse ALL cell models. 210 of the 556 functional regulatory variants are found in accessible chromatin sites that were identified in an ALL cell line. Source data are provided in the Source Data file.

For additional validation using traditional luciferase reporter assays, we prioritized these 210 functional regulatory variants based on allele-specific effect size and selected high-ranking SNVs. Dual-luciferase reporter assays showed similar allele-specific changes in activity to that which was detected by MPRA for 7 SNVs tested (Supplementary Fig.4ak). In fact, a significant positive correlation (p=0.0017) was observed between the allelic effects detected by MPRA and luciferase reporter assays (Supplementary Fig.4l). Together, these analyses assessed the robustness of our MPRA screen of functional regulatory variants and identified 556 SNVs with reproducible and concordant allele-specific effects on gene regulation. Importantly, 210 of the 556 significant hits that were concordant in at least three cell lines were found in open chromatin sites in ALL cell lines and, therefore, warranted further exploration.

To better understand how these variants impact cellular phenotypes, we first determined if the 210 functional regulatory variants found in accessible chromatin sites in ALL cell lines could be directly associated with a target gene. While 35 functional regulatory variants were localized close (2.5kb) to nearby promoters (Fig.4a, Supplementary Data4 and 6), 175 variants were promoter-distal (>2.5kb), and therefore likely to map to CREs with unclear gene targets (Fig.4a). While CREs are often associated with the nearest genes, 3D chromatin looping methods are a more reliable method to associate a CRE with its target gene promoter. In pursuit of evidence-based association of promoters and specific CREs, we performed two related chromatin looping methods, H3K27Ac HiChIP59 and promoter capture HiC (CHiC)39, in 8 of 10 ALL cell lines used in MPRA and determined that 19 of the 175 non-promoter functional regulatory variants showed connectivity to distal promoters in the same cell line where allele-specific MPRA activity and chromatin accessibility were detected (Fig.4a, Supplementary Data6). Interestingly, H3K27Ac HiChIP and promoter CHiC called similar numbers of loops across all 8 cell lines (690,579 versus 660,313, respectively), but promoter CHiC loop calling was more consistent per cell line (Supplementary Fig.5, Supplementary Data7). HiChIP detected no looping at any of the 556 reproducible and concordant SNVs from the MPRA, and the 19 SNVs showing connectivity to a promoter were solely detected by promoter CHiC, further highlighting the utility of this method in GWAS-oriented studies41,60,61,62,63.

a Data show the number of functional regulatory variants mapping to open chromatin in cell lines that associate directly with promoters (within 2.5kb) or that are distally promoter-connected via promoter CHiC. b MPRA data show distal regulatory variants in accessible chromatin (some promoter-connected by promoter CHiC data) exhibit stronger effects on allele-specific activity than promoter-associated functional regulatory variants. ANOVA with KruskalWallis test was performed with Dunns correction for multiple comparisons. c Amongst distally promoter-connected functional regulatory, variants that map to intronic and distal intergenic sequences showed greater activity than those in UTRs. ANOVA with KruskalWallis test was performed with Dunns correction for multiple comparisons. d, e Data show the ranked allele-specific activity distribution of MPRA data for d promoter-associated functional regulatory variants and e distally promoter-connected functional regulatory variants. All source data and statistical parameters are provided in the Source Data file.

In prioritizing functional regulatory variants, we were interested in the gene regulatory impact of variants at TSS-proximal promoter-associated versus TSS-distal promoter-connected CREs as measured by MPRA. Interestingly, we found that SNVs found at TSS-distal open chromatin sites, promoter-associated or not, showed higher allele-specific changes in MPRA activity than those at promoters (Fig.4b). While we acknowledge that many of the 156 variants for which we did not detect a relationship with a promoter are likely to have meaningful gene targets, we focused on CREs containing variants with known gene targets in ALL cells for functional validation. Amongst the TSS-distal promoter-connected functional regulatory variants, we found that distal intergenic and intronic SNVs showed significantly higher allele-specific activity than those in UTRs (Fig.4c). These data suggest that the most robust allelic effects attributable to these regulatory variants are likely to occur at distal intergenic and intronic sites >2.5kb from the TSS of the target gene.

Next, we ranked TSS-proximal promoter-associated and TSS-distal promoter-connected functional regulatory variants by the geometric mean of their significant MPRA data to account for the magnitude of allele-specific activity and the reproducibility of a significant change across ALL cell lines (Fig.4d, e). This analysis identified rs1247117 as the most robust functional regulatory variants, which we then pursued for mechanistic understanding (Fig.4e).

We pursued functional validation of rs1247117 based on its highest-ranking geometric mean of MPRA allelic effect. rs1247117 is in high LD with two GWAS sentinel variants (rs1312895, r2=0.99; rs1247118, r2=1) that are associated with persistence of MRD after induction chemotherapy3. This functional regulatory variant maps to a distal intergenic region harboring chromatin accessibility downstream of the CACUL1 gene, for which it is an eQTL in EBV-transformed lymphocytes37. However, we found that rs1247117 loops to the EIF3A promoter in Nalm6 B-ALL cells (Fig.5a). We, therefore, explored how this accessible chromatin site might recruit transcriptional regulators that would depend on the allele present at rs1247117. For this, we first performed ChIP-seq for RNA pol II and H3K27Ac, which confirmed RNA Pol II occupancy and H3K27Ac enrichment in Nalm6 cells, indicating that rs1247117 is associated with an active CRE (Fig.5a). Through an examination of the underlying DNA sequence spanning rs1247117, we found that the reference guanine (G) risk allele at rs1247117 resides in a PU.1 transcription factor binding motif that is disrupted by the alternative adenine (A) allele (Fig.5b). Although the risk G allele is the reference allele, the alternative A allele is more common in human populations. Supporting PU.1 binding at this location, accessible chromatin profiling in primary ALL cells identified an accessible chromatin site and PU.1 footprint spanning rs1247117 in diverse ALL samples (Supplementary Fig.6a, b). Significantly greater chromatin accessibility at rs1247117 was also observed in heterozygous (GA) patient samples compared to patient samples homozygous for the alternative A allele (Supplementary Fig.6c), and the G allele at rs1247117 harbored significantly greater ATAC-seq read count compared to the A allele (Supplementary Fig.6d). Importantly, we determined that PU.1 was bound at this site in Nalm6 cells using CUT and RUN64 (Fig.5a).

a IGV genome browser image in Nalm6 cells showing the genomic context, chromatin accessibility, and EIF3A promoter connectivity using promoter CHiC of the top functional regulatory variant, rs1247117, with the highest allele-specific MPRA activity. Genomic binding profiles are also shown for RNA polymerase II (RNA Pol2), histone H3 lysine 27 acetylation (H3K27Ac), and PU.1. b rs1247117 lies in a PU.1 binding motif. The human genome reference sequence, Nalm6 genome sequence, location of rs1247117, and PU.1 position weight matrix are shown. c Design of biotinylated DNA probes for in vitro rs1247117 pulldown. d Biotinylated DNA pulldown shows rs1247117 allele-dependent enrichment of PU.1 binding. Blot shown is representative of two independent experiments. Densitometric quantification of two blots is shown. e CRISPR/Cas9 was used to change the allele at rs1247117 from A>G in Nalm6 cells. Data show the location of gRNA and ssODN, as well as NGS reads obtained from clone 1 and 2 at rs1247117. f PU.1 ChIP-PCR shows increased PU.1 binding at the rs1247117 locus using two A>G modified clones and 3 primer sets. Data shown are meanSD of three independent experiments for each primer set. Two-way ANOVA with Dunnetts multiple comparisons correction, n=3. g ATAC-seq data normalized for frequency of reads in peaks (FRIP) show a significantly higher count of G nucleotides in two clones of A>G modified Nalm6 cells compared to the count of A nucleotides detected in parental Nalm6 cells. Data shown are the meanSD. Bonferroni corrected, two-tailed Students T tests, n=3. h Western blots and quantification showing decreased EIF3A expression in A>G modified Nalm6 cells. Blots shown are representative of three independent experiments. Quantification data shown are the meanSD. Two-tailed Students T tests compare parental Nalm6 to combined data from A>G clones, n=3. All source data and statistical parameters are provided in the Source Data file.

Nalm6 cells contain the alternative A allele that disrupts the PU.1 motif at rs1247117, yet our data suggests that this site still binds PU.1 (Fig.5a, b). This led us to hypothesize that PU.1 binding affinity for the PU.1 motif surrounding rs1247117 would be strengthened by the risk G allele. Therefore, we designed biotinylated DNA probes containing two tandem 25-bp regions centered on reference G or alternative A allele-containing rs1247117 to test this hypothesis (Fig.5c). Using biotinylated probes, we performed an in vitro DNA-affinity pulldown from Nalm6 nuclear lysate and found that while PU.1 was indeed bound to the alternative A allele, PU.1 was more robustly bound to the reference G allele at rs1247117 (Fig.5d). To further assess the impact of the rs1247117 allele on PU.1 binding, we changed the Nalm6 allele from A to G using CRISPR/Cas9 (Fig.5e; AA = parental genotype, GG = mutated genotype). We used ChIP-PCR to determine that PU.1 binding was increased with the G allele relative to the A allele at the CRE containing rs1247117 in two A>G Nalm6 clones across 3 unique primer sets within the PU.1 peak at rs1247117 that was detected in Nalm6 cells (Fig.5f). We then asked if transposase accessibility was also increased at the CRE containing rs1247117 when the G allele was present. Using ATAC-seq, we found that accessibility was indeed increased at rs1247117 in mutated Nalm6 cells with the G allele when compared to the parental Nalm6 cells containing the A allele (Fig.5g). These data suggest that the risk G allele increases genomic accessibility and the affinity of PU.1 binding at rs1247117 relative to the alternative A allele.

We were next interested in how allele-specific PU.1 binding at rs1247117 was related to the expression of the protein encoded by the connected gene, EIF3A. We found that the G allele, which increased recruitment of PU.1, resulted in decreased expression of EIF3A when compared to Nalm6 cells with the A allele (Fig.5h). These data suggest that PU.1 recruitment to the CRE containing rs1247117 results in a net-repressive effect on EIF3A protein levels, and that less PU.1 recruitment with the A allele results in greater EIF3A expression.

Clonal selection can lead to the accumulation of random SNVs and even larger structural variations65 that can confound functional interpretation of more complex trans phenotypic effects. Therefore, to examine the connection between rs1247117 and the persistence of MRD after induction chemotherapy, we decided to use CRISPR/Cas9 to delete the CRE containing rs1247117 in heterogeneous cell pools of Nalm6 and SUPB15 cells (rs1247117 del) to avoid clonal selection (Fig.6a, b, Supplementary Fig.7a). Given that loss of the CRE containing rs1247117 would abolish PU.1 recruitment at this region, we hypothesized that rs1247117 del would result in increased EIF3A expression. Accordingly, we found that EIF3A expression was elevated in rs1247117 del cells relative to parental Nalm6 and SUPB15 cells, respectively (Fig.6c, d, Supplementary Fig.7b), further supporting an inverse relationship between PU.1 binding at rs1247117 and EIF3A expression.

a Diagram on the left showing the genomic context of the rs1247117 CRE deletion in Nalm6 cells in relation to chromatin accessibility, PU.1 binding and rs1247117. Black bar represents ATAC-seq peak, green par represents PU.1 peak, and red bar represents region deleted using CRISPR/Cas9 genome editing. b Gel shows validation of deletion using primers flanking deleted region. Arrow points to PCR fragment with deletion in heterogeneous Nalm6 cell pools harboring deletion compared to wild-type parental Nalm6 cells. c EIF3A gene expression is upregulated upon deletion of the CRE containing rs1247117. RT-qPCR data show the meanSD of three independent experiments. Two-tailed Students T test. d Western blots and quantification showing increased EIF3A expression in rs1247117 del Nalm6 cells. Blots shown are representative of four independent experiments. Quantification data show the meanSD. Two-tailed Students T tests, n=4. eg Drug sensitivity data comparing viability relative to vehicle treatment of wild-type parental Nalm6 cells and Nalm6 cells with rs1247117 CRE deletion after vincristine (VCR) treatment for 24 (n=3), 48 (n=3) and 72 (n=3) hours at the indicated concentrations. Non-linear regression and F test analysis indicate that these dose-response curves are significantly different. h Caspase 3/7 activity assays comparing Caspase activity relative to vehicle treatment of wild-type parental Nalm6 cells and Nalm6 cells with rs1247117 CRE deletion after vincristine (VCR) treatment for 72hours at the indicated concentrations (n=3). Dose-response curves of non-linear regression indicate that these curves are significantly different. Non-linear regression and F test analysis indicate that these dose-response curves are significantly different. All source data are provided in the Source Data file.

Because the risk G allele at rs1247117 was also associated with vincristine resistance in primary ALL cells from patients, we additionally sought to determine the impact of the CRE deletion containing rs1247117 on cellular response to vincristine treatment. We hypothesized that because the risk G allele is associated with enhanced PU.1 binding and resistance to vincristine, complete disruption of PU.1 binding in Nalm6 cells harboring the CRE deletion would show increased sensitivity to vincristine relative to parental Nalm6 cells. As predicted, Nalm6 cells with the CRE deletion exhibited significantly increased sensitivity to vincristine across a range of concentrations after 24, 48, and 72hours of treatment (Fig.6eg), and we found consistent effects on cell viability in SUPB15 cells (Supplementary Fig.7c). Consistent with enhanced sensitivity to vincristine, we also found increased caspase 3/7 activity in rs1247117 del Nalm6 cells relative to parental Nalm6 cells after 72hrs and across a range of vincristine concentrations (Fig.6h). These data suggest that a functional regulatory variant alters the binding affinity of a key transcription factor, PU.1, and disruption of this locus impacts EIF3A expression and vincristine sensitivity in ALL cells. To further validate our methodology utilizing CRISPR/Cas9 to delete CREs, we deleted CREs spanning two additional top variants, rs7426865 and rs12660691 (see Fig.4e), that was associated with the ex vivo resistance to 6-mercaptopurine and dexamethasone, respectively, in primary ALL cells. Deletion of these CREs also impacted protein expression and sensitivity to the associated chemotherapeutic agent, thereby supporting our functional approach (Supplementary Figs.8 and 9).

We next wanted to connect EIF3A directly to vincristine resistance. Given that EIF3A is an essential gene per the Broad Institutes DepMap, we opted to test the hypothesis EIF3A overexpression alone was sufficient to impact the Nalm6 cell response to vincristine. We, therefore, used lentiviral transduction to overexpress EIF3A in Nalm6 cells and compared EIF3A overexpression (EIF3A OE) cells to control infected cells (Nalm6 WT, Supplementary Fig.10a). Using two independent infections of EIF3A OE, we found that at 48hr and 72hr, EIF3A OE cells were more sensitive to vincristine than Nalm6 WT cells (Supplementary Fig.10b). These data suggest that EIF3A expression impacts the ALL cell response to vincristine, with higher expression sensitizing cells to the drug, and further establishes this gene as the likely target of the association.

See the article here:
Investigation of inherited noncoding genetic variation impacting the pharmacogenomics of childhood acute ... - Nature.com

Recommendation and review posted by G. Smith

Pharmacogenomics could improve medication safety and reduce waste – Healthcare IT News

Posted: May 6, 2024 at 2:47 am

At present, pharmacogenomic tests are not available for all medications and are not widely employed as preventive measures in patient care. Globally, health insurance often does not even cover pharmacogenomic tests. This may change in the future, however, especially as pharmacogenomic testing becomes less expensive. Since an individual's genetic makeup remains constant, a pharmacogenomic test only needs to be performed once and bring lifelong benefits.

Challenges in broader adoption

There are several challenges to turning pharmacogenomictesting into routine practice:It would require investments in both technology and upskilling the workforce. Healthcare systems across the globe face the challenge of moving care upstream and moving to more preventative models of care,according to Videha Sharma, clinical innovation lead for the University of Manchester. "The prescribing of medicines is the most common therapeutic intervention in healthcare and offers a fantastic opportunity to avoid harmful side effects to make medicines more effective from the start. As such, there is a huge potential to boost the way we manage diseases at scale," Sharma said.

Current clinical use cases

Pharmacogenomics is gradually being introduced into clinical care, though it has not yet become a standard practice. In 2023, the National Institute for Health and Care Excellence (NICE) published draft guidance recommending point-of-care genomic testing for people who have had a stroke. The purpose of this test is to detect whether there have been changes in a gene called CYP2C19. This specific mutation can guide prescribing.

For example, in cardiology, patients with coronary artery disease, vascular diseaseor stroke are often prescribed a drug called clopidogrel. However, a patient may be a poor metabolizerof the drug, which CYP2C19 testing would revealin such cases, the patient would be offered an alternative.

Another example of whenpharmacogenomic testing is valuable is prior to administering the antibiotic gentamicin to infants, since one in 500 babies can suffer permanent hearing loss when prescribed this drug.This can be prevented by detecting the CYP2C19 mutation.

However, there are uncertainties around how to implement testing, how to share results across care settings and what the role of patients is so theyfeel empowered to receive personalised medicines. As a result, Sharma advocates for strong multi-disciplinary and cross-industry collaboration and has actively helped build a team of clinicians, designers, technologists and public contributors.

Upcoming plans of the NHS

In 2022, the British Pharmacological Society and the Royal College of Physicians published a report that calls for pharmacogenomic testing to be integrated fully, fairly and swiftly into the NHS in the UK. According to the authors, this will empower healthcare professionals to deliver better, more personalised care, and in turn improve outcomes for patients and reduce costs to the NHS.

The desire to advance pharmacogenomics in the clinical practice is there; it will simply require some time to achieve this goal. The "what" and the "why" have been clearly stated and are obvious to most key stakeholders the question of "how"still remains, and bridging the gap, genomics and digital health together will help realise the benefits of pharmacogenomics to patients and populations.

Clinical Innovation Lead for the University of Manchester Videha Sharma will be speaking at the Precision Digital Solutions for Personalised Care session during the 2024 HIMSS European Health Conference & Exhibition, which is scheduled to take place 29-31 May2024in Rome. Learn more and register.

Read this article:
Pharmacogenomics could improve medication safety and reduce waste - Healthcare IT News

Recommendation and review posted by G. Smith

Apples AI research suggests features are coming for Siri, artists, and more. – The Verge

Posted: May 6, 2024 at 2:47 am

It would be easy to think that Apple is late to the game on AI. Since late 2022, when ChatGPT took the world by storm, most of Apples competitors have fallen over themselves to catch up. While Apple has certainly talked about AI and even released some products with AI in mind, it seemed to be dipping a toe in rather than diving in headfirst.

But over the last few months, rumors and reports have suggested that Apple has, in fact, just been biding its time, waiting to make its move.There have been reports in recent weeks that Apple is talking to both OpenAI and Google about powering some of its AI features, and the company has also been working on its own model, called Ajax.

If you look through Apples published AI research, a picture starts to develop of how Apples approach to AI might come to life. Now, obviously, making product assumptions based on research papers is a deeply inexact science the line from research to store shelves is windy and full of potholes. But you can at least get a sense of what the company is thinking about and how its AI features might work when Apple starts to talk about them at its annual developer conference, WWDC, in June.

I suspect you and I are hoping for the same thing here: Better Siri. And it looks very much like Better Siri is coming! Theres an assumption in a lot of Apples research (and in a lot of the tech industry, the world, and everywhere)that large language models will immediately make virtual assistants better and smarter. For Apple, getting to Better Siri means making those models as fast as possible and making sure theyre everywhere.

In iOS 18, Apple plans to have all its AI features running on an on-device, fully offline model, Bloomberg recently reported. Its tough to build a good multipurpose model even when you have a network of data centers and thousands of state-of-the-art GPUs its drastically harder to do it with only the guts inside your smartphone. So Apples having to get creative.

In a paper called LLM in a flash: Efficient Large Language Model Inference with Limited Memory (all these papers have really boring titles but are really interesting, I promise!), researchers devised a system for storing a models data, which is usually stored on your devices RAM, on the SSD instead. We have demonstrated the ability to run LLMs up to twice the size of available DRAM [on the SSD], the researchers wrote, achieving an acceleration in inference speed by 4-5x compared to traditional loading methods in CPU, and 20-25x in GPU. By taking advantage of the most inexpensive and available storage on your device, they found, the models can run faster and more efficiently.

Apples researchers also created a system called EELBERT that can essentially compress an LLM into a much smaller size without making it meaningfully worse. Their compressed take on Googles Bert model was 15 times smaller only 1.2 megabytes and saw only a 4 percent reduction in quality. It did come with some latency tradeoffs, though.

In general, Apple is pushing to solve a core tension in the model world: the bigger a model gets, the better and more useful it can be, but also the more unwieldy, power-hungry, and slow it can become. Like so many others, the company is trying to find the right balance between all those things while also looking for a way to have it all.

A lot of what we talk about when we talk about AI products is virtual assistants assistants that know things, that can remind us of things, that can answer questions, and get stuff done on our behalf. So its not exactly shocking that a lot of Apples AI research boils down to a single question: what if Siri was really, really, really good?

A group of Apple researchers has been working on a way to use Siri without needing to use a wake word at all; instead of listening for Hey Siri or Siri, the device might be able to simply intuit whether youre talking to it. This problem is significantly more challenging than voice trigger detection, the researchers did acknowledge, since there might not be a leading trigger phrase that marks the beginning of a voice command. That might be why another group of researchers developed a system to more accurately detect wake words. Another paper trained a model to better understand rare words, which are often not well understood by assistants.

In both cases, the appeal of an LLM is that it can, in theory, process much more information much more quickly. In the wake-word paper, for instance, the researchers found that by not trying to discard all unnecessary sound but, instead, feeding it all to the model and letting it process what does and doesnt matter, the wake word worked far more reliably.

Once Siri hears you, Apples doing a bunch of work to make sure it understands and communicates better. In one paper, it developed a system called STEER (which stands for Semantic Turn Extension-Expansion Recognition, so well go with STEER) that aims to improve your back-and-forth communication with an assistant by trying to figure out when youre asking a follow-up question and when youre asking a new one. In another, it uses LLMs to better understand ambiguous queries to figure out what you mean no matter how you say it. In uncertain circumstances, they wrote, intelligent conversational agents may need to take the initiative to reduce their uncertainty by asking good questions proactively, thereby solving problems more effectively. Another paper aims to help with that, too: researchers used LLMs to make assistants less verbose and more understandable when theyre generating answers.

Whenever Apple does talk publicly about AI, it tends to focus less on raw technological might and more on the day-to-day stuff AI can actually do for you. So, while theres a lot of focus on Siri especially as Apple looks to compete with devices like the Humane AI Pin, the Rabbit R1, and Googles ongoing smashing of Gemini into all of Android there are plenty of other ways Apple seems to see AI being useful.

One obvious place for Apple to focus is on health: LLMs could, in theory, help wade through the oceans of biometric data collected by your various devices and help you make sense of it all. So, Apple has been researching how to collect and collate all of your motion data, how to use gait recognition and your headphones to identify you, and how to track and understand your heart rate data. Apple also created and released the largest multi-device multi-location sensor-based human activity dataset available after collecting data from 50 participants with multiple on-body sensors.

Apple also seems to imagine AI as a creative tool. For one paper, researchers interviewed a bunch of animators, designers, and engineers and built a system called Keyframer that enable[s] users to iteratively construct and refine generated designs. Instead of typing in a prompt and getting an image, then typing another prompt to get another image, you start with a prompt but then get a toolkit to tweak and refine parts of the image to your liking. You could imagine this kind of back-and-forth artistic process showing up anywhere from the Memoji creator to some of Apples more professional artistic tools.

In another paper, Apple describes a tool called MGIE that lets you edit an image just by describing the edits you want to make. (Make the sky more blue, make my face less weird, add some rocks, that sort of thing.) Instead of brief but ambiguous guidance, MGIE derives explicit visual-aware intention and leads to reasonable image editing, the researchers wrote. Its initial experiments werent perfect, but they were impressive.

We might even get some AI in Apple Music: for a paper called Resource-constrained Stereo Singing Voice Cancellation, researchers explored ways to separate voices from instruments in songs which could come in handy if Apple wants to give people tools to, say, remix songs the way you can on TikTok or Instagram.

Over time, Id bet this is the kind of stuff youll see Apple lean into, especially on iOS. Some of it Apple will build into its own apps; some it will offer to third-party developers as APIs. (The recent Journaling Suggestions feature is probably a good guide to how that might work.) Apple has always trumpeted its hardware capabilities, particularly compared to your average Android device; pairing all that horsepower with on-device, privacy-focused AI could be a big differentiator.

But if you want to see the biggest, most ambitious AI thing going at Apple, you need to know about Ferret. Ferret is a multi-modal large language model that can take instructions, focus on something specific youve circled or otherwise selected, and understand the world around it. Its designed for the now-normal AI use case of asking a device about the world around you, but it might also be able to understand whats on your screen. In the Ferret paper, researchers show that it could help you navigate apps, answer questions about App Store ratings, describe what youre looking at, and more. This has really exciting implications for accessibility but could also completely change the way you use your phone and your Vision Pro and / or smart glasses someday.

Were getting way ahead of ourselves here, but you can imagine how this would work with some of the other stuff Apple is working on. A Siri that can understand what you want, paired with a device that can see and understand everything thats happening on your display, is a phone that can literally use itself. Apple wouldnt need deep integrations with everything; it could simply run the apps and tap the right buttons automatically.

Again, all this is just research, and for all of it to work well starting this spring would be a legitimately unheard-of technical achievement. (I mean, youve tried chatbots you know theyre not great.) But Id bet you anything were going to get some big AI announcements at WWDC. Apple CEO Tim Cook even teased as much in February, and basically promised it on this weeks earnings call. And two things are very clear: Apple is very much in the AI race, and it might amount to a total overhaul of the iPhone. Heck, you might even start willingly using Siri! And that would be quite the accomplishment.

Original post:

Apples AI research suggests features are coming for Siri, artists, and more. - The Verge

Recommendation and review posted by G. Smith


Page 11234..1020..»