Annexon Announces Pricing of $125.0 Million Underwritten Public Offering of Common Stock

BRISBANE, Calif., Dec. 20, 2023 (GLOBE NEWSWIRE) -- Annexon, Inc. (NASDAQ: ANNX), a clinical-stage biopharmaceutical company developing a new class of complement medicines for patients with classical complement-mediated autoimmune, neurodegenerative and ophthalmic disorders, today announced the pricing of an underwritten public offering of 25,035,000 shares of its common stock and pre-funded warrants to purchase 18,379,861 shares of common stock. The shares of common stock are being sold at a price of $2.88 per share and the pre-funded warrants are being sold at a price of $2.879 per share.

Follow this link:
Annexon Announces Pricing of $125.0 Million Underwritten Public Offering of Common Stock

Alterity Therapeutics Appoints Phillip Hains as Chief Financial Officer

MELBOURNE, Australia and SAN FRANCISCO, Dec. 21, 2023 (GLOBE NEWSWIRE) -- Alterity Therapeutics (ASX: ATH, NASDAQ: ATHE) (“Alterity” or “the Company”), a biotechnology company dedicated to developing disease modifying treatments for neurodegenerative diseases, today announced the appointment of Phillip Hains as the Company’s new Chief Financial Officer (CFO), effective 31 January 2024.

Continue reading here:
Alterity Therapeutics Appoints Phillip Hains as Chief Financial Officer

Stem cell technology developed at UWMadison leads to new understanding of Autism risks – University of Wisconsin-Madison

RosetteArrays, developed at UWMadison, grow fields of neural rosettes embroynic versions of down-scaled, simplified brain structures from stem cells, giving scientists the opportunity to study the development of disorders like autism, spina bifida and epilepsy. Image courtesy of Neurosetta

Technology developed at the University of WisconsinMadison to grow rosettes of brain and spinal tissue gives scientists new ways to study the growing human brain, including a recent study of how genetic mutations linked to autism affect early stages of human brain development.

Its the latest discovery using RosetteArray technology, a screening tool that uses stem cells to generate embryonic forebrain or spinal cord tissue structures called neural rosettes. Neural rosettes are the starting material for generating human stem cell-derived neural organoids clusters of cells that resemble larger, more complex organs and can be used to assess whether different genetic makeups or exposure to chemicals increase the risk of neurodevelopmental disruptions.

Randolph Ashton

This technology gives us access to an embryonic model of human central nervous system development that we would otherwise not have access to, says Randolph Ashton, a UWMadison professor of biomedical engineering and associate director of the Stem Cell and Regenerative Medicine Center. This is useful, because not only can we now understand more about human development, but we can get an understanding of when it goes wrong.

Ashton and Gavin Knight, a scientist at the Wisconsin Institute for Discovery who earned his doctorate in Asthons lab, developed the technology behind RosetteArrays, which are marketed by Neurosetta, a company they co-founded with support from UWMadison Discovery to Product and the Wisconsin Alumni Research Foundations (WARF) Accelerator Program.

RosetteArray technology played an important role in a study published recently in Nature Neuroscience. The study, led by University of Southern California stem cell biologist Giorgia Quadrato, with Ashton and Knight as co-authors, investigated mutations of a gene called SYNGAP1.

SYNGAP1 mutations have long been associated with risk factors for autism spectrum disorder, epilepsy, neurodevelopmental disability and more, but until now the gene has mainly been studied in animal models and focued on the impact of SYNGAP1 on synapses, the structure at the tips of long brain cells called neurons that allow them to pass signals to neighboring cells.

In their new SYNGAP1 autism study, Quadrato and her lab used RosetteArray technology to grow neural rosettes from healthy human cells as well as from the cells of a patient with a disease-causing variant in SYNGAP1. By analyzing these young, developing neural organoids, Quadrato determined that human radial glia cells the cells responsible for producing all the neurons in the outer layer of the brain called the cerebral cortex can express SYNGAP1. When SYNGAP1 is mutated, it leads to disrupted organization of the cortical plate, an early brain structure that gives rise to the cerebral cortex. This shows that SYNGAP1-related brain disorders can arise through non-synaptic mechanisms.

Quadrato Lab and Neurosetta plan to partner on further studies to explore the extent of autism spectrum disorder genetic backgrounds that can be modeled using RosetteArray technology, which Ashton hopes will eventually lead to new precision medicine approaches.

Simply being able to model early human development, in this case brain and spinal cord formation, gives you a very powerful platform to try to improve human health, says Ashton. Weve been surprised to see the effects of neurological disease-causing mutations in the earliest stages of these tissues formation. RosetteArrays model approximately four to six weeks post conception, and were learning that you can start to see markers for autism then, which is a disease that people typically arent diagnosed with until post 2 years of age. So, the fact that we can see this very early in our model of human development is amazing.

Ashton says researchers using technologies like the RosetteArray are finding that the risk factors for autism spectrum disorder are boiling down to a couple of core pathways, that seem to have roles very early in human brain development, which is helpful information as researchers work on treatments.

While this paper focused on studying brain tissue, Ashton has used the RosetteArray platform in his own lab to study defects in neural tube formation.

(The neural tube) is a structure that goes from the head of the embryo all the way down through the back of the spinal cord. All brain, spinal cord and eye tissue comes from this neural tube, says Ashton. It so happens that a lot of things can disrupt that process, and if that formation is disrupted early enough, then it causes lots of issues. It can cause congenital birth defects known as neural tube defects, for example spinal bifida, which is when the lower spine doesnt fully close. Or, if you have a failure of closure higher in the neural tube that leads to a failed pregnancy, so understanding this process is crucial.

Ashton and his lab members have been using RosetteArrays to investigate what may be causing spina bifida defects and how they can be mitigated.

There are examples of known chemicals we use in our food supply, pesticides, and anti-cancer drugs that have historically been correlated with causing neural tube defects. So, its important that we have a way to test new chemistries and chemical processes to make sure they dont have these effects on human development, says Ashton. Weve used rodent models but theres a difference between animals and humans. The RosetteArray provides a way to test these chemicals on early human brain and spinal cord development.

The RosetteArray platform may also be used for individualized medicine, as it can be used to screen individual patients cell lines to better understand how mutations in a persons genomic background can lead to a disorder as well as how the interaction between a persons genomic background and the chemicals that theyre exposed to may lead to a health risk.

We think this platform will be highly useful for both commercial applications for screening for chemicals that can cause neurodevelopmental risk, as well as for clinical application, Ashton says. And I think the real power of the tool is for precision medicine and drug discovery.

Follow this link:
Stem cell technology developed at UWMadison leads to new understanding of Autism risks - University of Wisconsin-Madison

Casgevy and Lyfgenia: Two Gene Therapies Approved for Sickle Cell Disease – Yale Medicine

Until recently, the only possible cure for sickle cell disease, an inherited genetic blood disorder most common in people with African ancestry, was a bone marrow transplant, which has its own set of challenges.

Now, people with sickle cell disease (SCD)which affects an estimated 100,000 Americans and can cause chronic pain, organ damage, strokes, and shortened life expectancyhave additional, potentially curative options. In early December, the Food and Drug Administration (FDA) approved two gene therapies for SCD, one of which is the first approved medication that uses the gene-editing tool CRISPR.

Both treatmentsCasgevy, which is made by Vertex Pharmaceuticals and CRISPR Therapeutics, and Lyfgenia, by Bluebird Bioare for people 12 and older. Sickle cell disease is a red blood cell disorder that affects hemoglobin, the protein that carries oxygen throughout the body. These two therapies work in different ways, but both are intended to be a one-time fix, although that will require years of follow-up to know for sure.

With Casgevy, an edit (or cut) is made in a particular gene to reactivate the production of fetal hemoglobin, which dilutes the faulty red blood cells caused by sickle cell disease (more on that below). Lyfgenia, on the other hand, uses a viral envelope to deliver a healthy hemoglobin-producing gene.

The therapies are hailed as groundbreaking as they represent the first-ever gene therapies to potentially cure a hereditary condition.

For many years, we only had one treatment for sickle cell disease, and then medicine advanced to the point where we could offer bone marrow transplant, the first potential cure for sickle cell disease, says Cece Calhoun, MD, MBA, a Yale Medicine hematologist-oncologist. But trying to find a good match for a transplant is a big barrier. This new technology uses gene therapy to allow patients to be their own match.

This is significant, she notes, because a sickle cell crisisthe pain the disease causesis unpredictable and intense, akin to how it feels to have a long bone fracture.

But, says Dr. Calhoun, the pain sickle cell disease causes is not the only problem faced by people with the condition.

Sickle cell disease impacts every organ. Children are having strokes, and young adultspeople in their 30sare experiencing kidney failureall because of sickle cell disease. If we can intervene and prevent these complications and let these patients live full lives, that is huge, Dr. Calhoun says.

Lakshmanan Krishnamurti, MD, chief of Yale Medicine Pediatric Hematology & Oncology, agrees.

Many cant have bone marrow transplantation because only about 15% of patients have a matched sibling, and we can find an unrelated donor for only another 10% to 12%. That means we are only helping 25% of patients, says Dr. Krishnamurti, who was an author on the Lyfgenia study published in The New England Journal of Medicine. This is a big step forward.

However, the gene therapies are time-intensivetaking about a year to complete the processand grueling. As with bone marrow transplants, they require high-dose chemotherapy to kill the faulty stem cells before they are replaced with modified stem cells.

The gene therapies will be available only at large, authorized medical centers because they require advanced care. They are also expensive (estimates put it at $2 to $3 million per patient), and its yet to be determined if or how insurance companies, including Medicaid, will cover the treatment.

Dr. Krishnamurti says both treatments will be available at Yale and that anyone interested in learning more should speak to their physician.

Below, Drs. Calhoun and Krishnamurti answer common questions about sickle cell disease and these new gene therapies.

See the original post here:
Casgevy and Lyfgenia: Two Gene Therapies Approved for Sickle Cell Disease - Yale Medicine

Paralyzed B.C. man has hopes in stem cell treatment for recovery journey – Global News

A paralyzed B.C. man has hopes that stem cell treatment will help him regain sensation and movement in his body.

On July 21, 2021, Cameron Thompson, 25, was swimming at Puntledge River in Courtenay, B.C., on Vancouver Island. He dove into the water head first and ended up severing his spinal cord.

That day when I dove into the water, I knew instantly that I had become paralyzed, he told Global News.

I just thought, I dont want to choke on water so I just held my breath thinking I would hold it until I passed out.

Thompsons friends then were able to pull him out of the water and called 911.

I remember everything until I got into the ambulance, he said. And then I woke up the next day in Vancouver.

Story continues below advertisement

Thompson had lost the majority of the functions of his body, except partial movement to his shoulders and arms.

Super thankful for my friends without them I would have floated down the river, Thompson said.

The Thompson family has created an online fundraiser, hoping to raise $50,000 for stem cell treatment at BioXcellerator.

According to BioXcellerator, the company is recognized as a global leader in treating a wide range of conditions based on 25 years of clinically-based research with stem cell therapy.

2:00 B.C. communities rank high on list of Canadas most generous cities

With the stem cell treatment, Thompson hopes to reduce his nerve pain which will boost his immune system and could help him regain sensation and movement in his body.

Thomson said his recovery journey so far has been a tough road to travel but he has not let the injury affect his mindset and outlook on life.

Story continues below advertisement

I have the same outlook on life I have hard days thinking about what life could have been. But, day by day, things get easier. I understand there will always be struggles with life. Other people have it worse so you just deal with what you got, he said.

Trending Now

In terms of a possible future career, Thompson said he is focusing on investments and the stock markets.

I want other people that have spinal injuries to know there are things that can help them and there are people that understand what they are going through, Thompson said.

Reaching out and talking can make a really big difference.

Global News spoke with Camerons mother who said the road has been tough, but she is very hopeful for the stem cell treatment.

Even if it partly works, it will be amazing, said Christina Thompson

Its a blessing. The fundraiser is just for the treatment. If we could raise the money it would be a godsend.

She said none of the costs would be covered by the government.

1:42 Friends and family set up fundraiser for Pitt Meadows boy killed in crash

2023 Global News, a division of Corus Entertainment Inc.

See more here:
Paralyzed B.C. man has hopes in stem cell treatment for recovery journey - Global News

Embryonic-stem-cell-derived mesenchymal stem cells relieve experimental contact urticaria by regulating the functions … – Nature.com

Animals

All animal experimental procedures were approved by the Dong-A University Medical School Institutional Animal Care and Use Committee (Approval No. DIACUC-21-11) at 22.03.2021. Female BALB/c mice that were 78weeks old were purchased from Orient Bio Inc. (Gyeonggi-do, korea). The classification of experimental groups involves the random assignment of mice of the same age and within a 1g difference in body weight after the acclimatization process. The mice are then housed in a pathogen-free facility at Dong-A University (Busan, Korea). Mice were maintained at Dong-A University facility at 22C1C room temperature, 4060% humidity, on a 12h lightdark cycle (7a.m. to 7p.m.), and given food and water freely, according to institutional guidelines. All experiments were performed under inhalation anesthesia with isoflurane, and mice were euthanized by CO2 inhalation at the end of the experiment. This study adhered to the guidelines set forth by the laboratory animal ethics committee of Dong-A University and the ARRIVE guidelines. To ensure statistical significance, 5 or more mice per group were used, and all experimental protocols were approved by the Institutional Animal Care and Use Committee (IACUC) of Dong-A University. Inhalational anesthesia using isoflurane was used to induce anesthesia when sacrificing all experimental animals.

Contact urticaria mouse model was induced according to a previously reported method31,32. Mice were initially sensitized by applying 100l of a TMA (trimellitic anhydride; 500mg/ml, Alfa Aesar, Ward Hill, MA, USA) in acetone/olive oil (4:1, v/v) on the shaved hind flank. This sensitization process is essential for inducing an immune response to TMA. Secondary sensitizations were performed on the hind flank to reinforce the immune response. On the 7th and 10th days after the first sensitization, mice sensitized 50l of a TMA solution (250mg/ml) in acetone/olive oil (4:1, v/v). On day 13 after the initial sensitization, contact urticaria (CU) was induced by challenging the ears with 25l of a TMA solution (100mg/ml) dissolved in acetone/olive oil (4:1, v/v). The disease symptoms were assessed by measuring ear thickness, itching, and lesions on the skin. Particularly, skin lesions were evaluated by determining the ratio of the affected area, indicating erythema and edema on a 3 cm2 area of the dorsal skin. The symptom evaluation of experimental animals was evaluated based on all animals without exclusion criteria. For histological analysis, H&E and mast cell staining were conducted. Cellular and molecular analysis involved the use of flow cytometry to assess immune cell activity in mouse lymphoid organs, along with genetic analysis of the lesions. Experimental animals of 10 mice per group were performed by blindly selecting 56 mice for efficient handling of animal-derived flow cytometric and genetic analysis. The corresponding author and one of the first authors (S.Y. Hyun) were aware of the group assignment, and the symptoms, flow cytometry, and other molecular analysis results were evaluated together by multiple blinded co-authors.

For the purposes of in vitro experiments, we used the nave CD3+ T cell isolation Kit (Miltenyi Biotec, Bergisch-Gladbach, Germany) to enrich nave CD3+ cells from the spleens of BALB/c mice (6weeks old). All steps were conducted strictly following the manufacturers protocol.

M-MSCs used in this study was provided Mirae Cell Bio (Seoul, Korea). M-MSCs differentiated from H9 hESCs30 were maintained in EGM2-MV medium (Lonza, San Diego, CA, USA) containing supplement Mix (promocell, Heidelberg, Germany) and 50 ug/ml Gentamicin (Gibco, NewYork, USA) in a humidified atmosphere containing 5% CO2 at 37, as previously described33. M-MSCs at less than ten passages were used for in vitro cell culture and in vivo animal experiments. Bone marrow-derived MSCs (BM-MSCs) were maintained in MSCBM medium (Lonza, Basel, Switzerland) containing supplement kit (Lonza) in a humidified incubator at 5% CO2 at 37. Bone marrow-derived mast cells (BMMCs) derived from BALB/c mice were cultured in RPMI 1640 medium containing 2mM L-glutamine, 0.1mM nonessential amino acids, antibiotics, 10% fetal bovine serum (FBS), and IL-3 (10ng/ml; PeproTech Inc., Rocky hill, NJ, USA). After 4weeks,>98% of the cells were verified as BMMCs, as previously described34. Mouse splenic T cells were presorted by CD3 mAb-microbeads (Miltenyi Biotec, Bergisch Gladbach, Germany) followed by the manufacturer's method. For T cell polarization, splenic nave CD3+ T cells were cultured onto a 24-well plate coated with 1g/ml of anti-CD3 (eBioscience, San Diego, CA, USA) in complete RPMI 1640 medium and supplemented with TH1 reagents [IL-2 (20ng/ml, PeproTech Inc.), IL-12 (20ng/ml, PeproTech Inc.), and anti-IL-4 (10g/ml, Bio X cell, West Lebanon, NH, USA)] or TH2 reagents [IL-2 (20ng/ml, PeproTech Inc.), IL-4 (100ng/ml, PeproTech Inc.), and anti-IFN- (10g/ml, Bio X cell)]. After 48h, the cells were co-cultured with M-MSCs for 24h under polarized conditions. To confirm the regulation of immune cells by M-MSCs in vitro, BMMCs (5.0105 cells/well), splenic T cells (5.0105 cells/well), or polarizing splenic T cells (5.0105 cells/well) were co-cultured with the indicated ratio of M-MSCs for 24h. Co-cultured splenic T cells were identified by flow cytometry analysis. The analysis of polarized T cells was assessed by distinguishing them using the gating strategy as depicted in Supplementary Fig.S1. The ratio of degranulation of BMMCs was analyzed by -hexosaminidase secretion.

After the primary sensitization of the contact urticaria model, M-MSCs were injected subcutaneously into the ear as a single administration on the 10th days or twice on the 10th and 12th days. A primary disease improvement evaluation was performed through single administration or two administrations, and an appropriate cell administration group for disease efficacy was selected. The administration of an equal amount of BM-MSCs and oral administration of cetirizine (50mg/kg) (Sigma-Aldrich, St. Louis, MO, USA) were used as positive controls. To deplete TGF-, BALB/c mice were intraperitoneally injected with 300g of anti-TGF- mAb (1D11.16.8, Bio X cell) or an isotype-matched control mAb (Bio X cell) twice on days 0 and 3 of M-MSCs administration.

Single-cell suspensions were isolated from the spleen, cervical lymph node (cLN), and ear. Ear tissues were isolated into single cells using the gentleMACS dissociator (Miltenyi Biotec) followed by the manufacturers method. For the detection of intracellular cytokines, the isolated cells were stimulated with PMA (50ng/ml; Sigma-Aldrich), ionomycin (500ng/ml; Sigma-Aldrich), and brefeldin A (3g/ml; eBioscience) for 4h before analysis and a fixation/permeabilization kit were from eBioscience. Before cell surface markers were stained, Fc receptors were blocked with anti-CD16 and anti-CD32 mAbs (2.4G2, BD Biosciences), and conjugated and dead cells were excluded by analysis on the basis of forward and side light scatter parameters and staining with a Zombie NIR Fixable Viability Kit (Biolegend, San Diego, CA, USA). The antibodies against proteins were as follows: Antibodies against CD3 (17A2) and CD8a (53-6.7) were obtained from BioLegend. Antibodies for CD4 (RM4-5), IFN- ((XMG1.2), IL-4 (11B11) were obtained from eBioscience. Antibodies for CD3 (17A2), CD45 (30-F11), and CD127 (A7R34) were obtained from BioLegend. The cells were then analyzed with a NovoCyte flow cytometer (Agilent) and FlowJo version 10 software (Tree Star, Ashland, OR, USA).

BMMCs (5.0105 cells/well) co-cultured with M-MSCs (0.5 to 2106 cells/well) for 24h were sensitized for 4h with Monoclonal dinitrophenol (DNP)-specific IgE (100ng/ml; Sigma). The IgE-primed BMMCs were then stimulated with 50ng/ml of DNP-human serum albumin (DNP-HSA, Sigma-Aldrich) in Tyrode-BSA buffer (20mM Hepes (pH 7.4), 135mM NaCl, 5mM KCl, 1.8mM CaCl2, 1mM MgCl2, 5.6mM glucose, and 0.1% BSA) for 15min in the presence or absence of the M-MSCs.

Degranulation was determined by measuring the release of the granule marker -hexosaminidase as previously described35. The degree of degranulation of BMMCs was expressed as the % of the activity of -hexosaminidase secreted out of the cells compared to the total activity of -hexosaminidase.

M-MSCs were co-cultured with splenic T cells or BMMCs for 24h and then effector cells were removed. M-MSCs were rinsed with PBS and left on ice for 5min to stop the reaction. Total RNA was extracted using AccuPrep Universal RNA Extraction Kit (Bioneer, Daejeon, Korea), and cDNA was synthesized using AccuPower CycleScript RT PreMix (Bioneer) according to the manufacturers instructions. The PCR reaction was amplified using AccuPower PCR PreMix (Bioneer) and PCR was performed at 95 for 2min, 95 for 20s, 58 for 40s, 72 for 30s, 72 for 5min for 30 cycles. Primers used as follow: human Hgf (forward 5-TCCATGATACCACACGAACACAGC-3, reverse 5-TGCACAGTACTCCCAGCGGGTGTG-3); human Ido1 (forward 5-TTTGCTAAAGGCGCTGTTGG-3, reverse 5-CCTTCATACACCAGACCGTCTGA-3); human Pdl1 (forward 5-TATGGTGGTGCCGACTACAA-3, reverse 5-TGCTTGTCCAGATGACTTCG-3); human Il10 (forward 5-AGACATCAGGGTGGCGACTCTAT-3, reverse 5-GGCTCCCTGGTTTCTCTTCCTAAG-3); human Pge2 (forward 5-ACCATCTACCCCTTCCTTT-3, reverse 5-CCGCTTCCCAGAGGATCT-3); human Tgfb (forward 5-GGGACTATCCACCTGCAAGA -3, reverse 5-CCTCTTGGCGTAGTAGTCG-3); human Gapdh (forward 5-ACCACAGTCCATGCCATCAC-3, reverse 5-TCCACCACCCTGTTGCTGTA-3). Snap-frozen disease-inducing mouse ear tissues were ground to powder. Total RNA isolation and PCR reaction were performed in the same manner as above. Real-time PCR was performed Thermal Cycler Dice Real Time System III TP950 (Takara, Shiga-ken, Japan). Primers used as follow: mouse Il4 (forward 5-ACAGGAGAAGGGACGCCAT-3, reverse 5-GAAGCCCTACAGACGAGCTCA-3); mouse Il6 (forward 5-GAGGATACCACTCCCAACAGACC-3, reverse 5-AAGTGCATCATCGTTGTTCATACA-3); mouse Ifng (forward 5-CAGCAACAGCAAGGCGAAAAAGG-3, reverse 5-TTTCCGCTTCCTGAGGCTGGAT-3); mouse Tnfa (forward 5-AGTGACAAGCCTGTAGCCCACGT -3, reverse 5-CCATCGGCTGGCACCACTAGTT-3); mouse Gapdh (forward 5-CATCACTGCCACCCAGAAGACTG-3, reverse 5-ATGCCAGTGAGCTTCCCGTTCAG-3);

After the induction of contact urticaria in mice, their ear tissues were fixed in 4% paraformaldehyde in phosphate-buffered saline for 24h and then embedded in paraffin. The tissues were dehydrated in a graded ethanol series (70 to 100%), rinsed three times with xylene for 3min each, and then embedded in paraffin. Sections of paraffin-embedded tissues, with a thickness of 6m, were prepared and stained with hematoxylin (Sigma-Aldrich) and eosin (Sigma-Aldrich) to compare and analyze the degree of cell invasion and epidermal thickness in the tissue. Additionally, sections of tissues with a thickness of 6m were stained with a 1% toluidine blue (Sigma-Aldrich) solution to assess the number of infiltrating mast cells and the degree of degranulation.

The in vitro experiment was repeated three independent times, and the animal experiment was based on five or more animals per group, and if the results of the first experiment were insufficient, the significance was evaluated within a total of 10 animals in the group. The data are presented as the meanstandard error (SEM) from three or more independent experiments for in vitro experiments. Statistical analysis was done by unpaired Student's t-test. One-way analysis of variance (ANOVA) with Tukey's post hoc test was performed for multiple comparisons. Statistical significance (*P<0.05 and **P<0.01) was determined with Prism version 7.0 (GraphPad, San Diego, CA).

This study was approved by the Institutional Animal Care and Use Committee (IACUC) of Dong-A University(DIACUC-21-11). All animal experiments were performed in accordance with the guidelines and regulationsof the institutional guidelines.

Go here to read the rest:
Embryonic-stem-cell-derived mesenchymal stem cells relieve experimental contact urticaria by regulating the functions ... - Nature.com

Machine learning-based estimation of spatial gene expression pattern during ESC-derived retinal organoid … – Nature.com

CNN architecture and dataset for estimating spatial gene expression patterns

Our model utilizes a CNN that takes a phase-contrast image as input and estimates a fluorescent image as output (Fig.1A). The typical input to a CNN is a two-dimensional (2D) image. This 2D image is passed through several convolution layers, each followed by a nonlinear activation function. The training parameters correspond to the weights of these convolution kernels and the biases. Our network has a U-Net-like architecture27, which is an encoder-decoder structure with skip connections. The embedded features from the encoder are passed through the decoder, which consists of upsampling and convolution layers to increase the resolution of the intermediate feature maps to obtain a fluorescent image as output.

In our model, the ResNet5028 was used as the backbone of the encoder. The size of the input image for the ResNet50 is (3times Htimes W). To use the pre-trained model of the ResNet50, gray-scale phase-contrast images were replicated in the axis of the channel to create three-channel images. At the first layer, a convolution with stride 2 is applied to the input image to generate features of size (64times frac{H}{2}times frac{W}{2}). The ResNet50 has 4 residual blocks and the size of the output features of these blocks are (256times frac{H}{4}times frac{W}{4}), (512times frac{H}{8}times frac{W}{8}), (1024times frac{H}{16}times frac{W}{16}), and (2048times frac{H}{32}times frac{W}{32}), respectively. These features are then concatenated to the decoder to exploit multi-scale information. The output of the decoder is a fluorescent image of size (1times frac{H}{2}times frac{W}{2}). Note that each convolution layer has a batch normalization (BN) layer and a rectified linear unit (ReLU) activation function, except for the final convolution layer, which has a sigmoid activation function to constrain the range of the output values between 0 and 1.

The network was optimized by minimizing the training loss computed on the output and corresponding ground-truth fluorescent images. The combination of mean squared error (MSE) and cosine similarity, which captures structural patterns from the entire image, was used as the training loss.

To train, validate, and test our model, we cultured retinal organoids derived from mouse ESCs using the SFEBq method10. In this culture, a GFP gene was knocked-in under the promoter of a master gene of retinal differentiation, Rax. Using this method, we obtained a dataset of a pair of phase-contrast image and fluorescent image of Rax during retinal differentiation (Fig.1B). Images were captured for 96 organoids at 4.5, 5, 6, 7, and 8days after the start of SFEBq, where each sample was captured as 14 Z-stack images. This resulted in a total of (96times 5times 14=6720) image pairs were obtained. These image pairs were divided into 5880, 420, and 420 samples for training, validation, and test, respectively. 84, 6, and 6 organoids were used for training, validation, and test, respectively; thus, each organoid does not appear in the different datasets. For data augmentation, we randomly flipped the input images vertically and horizontally during training. While the image resolution of both phase-contrast and fluorescent images is (960times 720), the (512times 512) regions where organoids appear were extracted.

To demonstrate our model, we applied it to 420 samples of the test data. As a result, the proposed model successfully estimated the spatial expression patterns of Rax from phase-contrast images during retinal organoid development (Fig.2). During development, multiple optic vesicles are formed through large and complicated deformations (Fig.2A). This process begins with a spherical embryonic body, with some portions of the tissue surface evaginating outward to form hemispherical vesicles, i.e., optic vesicles. Importantly, the resulting morphology of retinal organoids, especially optic vesicles, varies widely29. This process is known to be governed by the expression of the Rax gene (Fig.2B). That is, the Rax gene is gradually expressed in several parts of the tissue surface, so-called eye field, where cells differentiate from neuroepithelium into several types of retinal cells.

Estimated spatial Rax expression patterns during retinal organoid development. (A) Phase-contrast images from day 4.5 to day 8. (B) Captured fluorescent images of Rax as ground-truths. (C) Estimated fluorescent images with our model. (D) Error maps between captured and estimated images. The error metric was a squared error. The organoids in (AD) are identical. Scale bars indicate 200m.

Our model successfully recapitulated the above features of Rax expression (Fig.2C), i.e., the Rax intensity was relatively low and homogenous at days 4.5, 5, 6, and gradually increased around the evaginated tissue regions at days 7 and 8. Remarkably, the regions of high Rax expression were accurately estimated even in organoids with various morphologies. On the other hand, as the Rax intensity increases, especially around the evaginated tissue regions, the error of the estimated image from the ground-truth image increases with time (Fig.2D).

To quantitatively evaluate the accuracy of the estimation, we statistically analyzed the estimation results at each stage. To clarify whether the model can estimate Rax intensity in both samples with high and low Rax expression, each of the ground-truth and estimated fluorescence images was divided into two categories by the coefficient of variation of the foreground pixels in a fluorescent image at day 8 (Fig.3A). The samples in each group were labeled as positive and negative, respectively. For each of these categories, the mean and coefficient of variation of the pixel values were calculated (Fig.3BE). In calculating these values, the phase-contrast images were binarized to obtain foreground and background masks, and then computed using only the foreground pixels and normalized to those of the background pixels.

Statistical analysis of fluorescence at each developmental stage for positive and negative samples. (A) Histogram of coefficient of variation for foreground pixel values of fluorescent images at day 8. (B, C) Means of pixel values in positive and negative samples at each stage for ground-truth (green bars) and estimated fluorescent images (blue bars), respectively. (D, E) Coefficients of variation in positive samples at each stage for both ground-truth (green bars) and estimated fluorescent images (red bars), respectively. (F, G) Plots of ground-truth and estimated pixel values in positive and negative samples at each stage, respectively. Errors are 0% and 25% on the solid and dotted black lines, respectively. Error bars in (BE) indicate standard deviations.

Positive samples showed a gradual increase in mean and intensity over the days passed (Fig.3B). The negative sample, on the other hand, showed relatively low values from the beginning and did not change significantly over the days (Fig.3C). Similarly, the coefficients of variation increased in the positive samples but not in the negative samples (Fig.3D,E). These results indicate that the model successfully estimates the feature of the spatial Rax expression patterns during retinal organoid development, i.e., positive samples gradually increase Rax expressions and their heterogeneity, but negative samples do not. The intensity of the estimated images is relatively lower than the intensity of the ground-truth images in the positive samples and vice versa in the negative samples.

To clarify whether the model is capable to estimate intermediate values of the Rax expression, we analyzed the correlations between ground-truth and estimated values on foreground pixels at each stage, respectively (Fig.3F,G). The results show that in the positive sample (Fig.3F), the distribution of intensities is initially concentrated at low intensities and gradually expands to high intensities as the day progresses, with a wide distribution from low to high intensities. Similarly, in the negative sample, the luminance distribution is initially concentrated at low intensities, but does not expand as much as in the positive sample (Fig.3G). These results indicate that the model successfully estimated the plausible values across all pixel intensities, demonstrating the capability of our method to infer intermediate levels of gene expression. Notably, predicting Rax expression in the organoids at later stages, such as day 8 in our experiments, becomes more feasible for the model due to their characteristic morphologies. These distinct morphologies provide features that can be efficiently extracted by the convolution operators of the model.

To determine whether the estimated Rax expression patterns correspond to tissue morphologies, we quantified the spatial distribution of Rax intensity and the mean curvature along the tissue contour around each optic vesicle (Fig.4). For this analysis, four typical optic vesicles were selected from the positive samples and their curvature and Rax distribution were quantified. In this analysis, tissue contours were extracted and the radius of a circle passing through three points on the tissue contour was calculated as the inverse of the curvature. Moreover, the Rax intensity was measured as the average value along the depth direction from the tissue contour.

Correlation analysis of spatial Rax expression patterns and optic-vesicle morphologies. (A) Phase-contrast images. (B) Captured fluorescent images of Rax as ground-truths. (C) Estimated fluorescent images with our model. (D) Mean curvatures as a function of the distance along the organoid contour. (E) Captured and estimated fluorescent intensities of Rax along the organoid contour. The organoids in (AC) are identical and captured on day 8. The mean curvatures and fluorescence in (D, E) are for the regions indicated by the red line starting from the red dot in (A). Scale bars indicate 200m.

Optic vesicles are hemispherical, with positive curvature at the distal portion and negative curvature at the root (Fig.4A,D). The Rax intensity is continuously distributed around each vesicle, being highest at the distal part and gradually decreasing toward the root (Fig.4B,E). Furthermore, because the test images were taken with a conventional fluorescence microscope, structures above and below the focal plane are included in each image. Therefore, although some images have multiple overlapping vesicles (e.g., samples iii and iv), the model successfully estimated the Rax intensity of the overlapping regions as well.

MSE is commonly used as the training loss for training regression models. In addition to MSE, this model also uses cosine similarity, which can capture structural patterns from the entire image. To analyze the effect of cosine similarity on the estimation accuracy, we tested the model with different weights for both error metrics (Fig.5). The trained models were evaluated with MSE for each test dataset on different days (Fig.5A). The results demonstrated that cosine similarity improved the estimation accuracy at the early and intermediate stages, such as from day 4.5 to day 6. At these stages, the intensity in the differentiated region is weak, making it difficult for the network to capture structural patterns using MSE alone. Cosine similarity, on the other hand, enabled the network to learn the patterns from the weak intensity by calculating the correlation between the normalized ground-truth and the estimated images (Fig.5B). Therefore, our model has the capability to achieve the best estimate at different stages with appropriate weight balancing.

Effects of the balance of training loss on estimation accuracy. (A) Mean squared error at each stage with different hyperparameters, where bold and underlined numbers stand for the best and second best results on each day, respectively. (B) Examples of estimated fluorescent images at days 6 and 8 with different hyperparameters. The MSE of each estimated image is described in the upper left. The results with the lowest MSEs are surrounded by the red boxes. Scale bars indicate 200m.

See original here:
Machine learning-based estimation of spatial gene expression pattern during ESC-derived retinal organoid ... - Nature.com

Crucial blood stem cell creation step found by ISU researchers – Tech Explorist

A microbial sensor, Nod1, identifies bacterial infections and aids in developing blood stem cells, offering valuable insights. Raquel Espin Palazons team at Iowa State University discovered this, potentially eliminating the need for bone marrow transplants.

Published in Nature Communications, the finding builds on Espin Palazons earlier work, revealing the role of inflammatory signals in the embryos early stages and activating Nod1 in embryos forces vascular cells to become blood stem cells. This knowledge could pave the way for creating patient-specific blood stem cells derived from their own blood in the lab.

Espin Palazon said, This would eliminate the challenging task of finding compatible bone marrow transplant donors and the complications that occur after a transplant, improving the lives of many leukemia, lymphoma, and anemia patients.

Stem cells act as both the builders and raw materials in our bodies, constantly dividing to renew and create cells for different tissues. Embryonic pluripotent stem cells can become any cell type, while adult stem cells are limited.

Blood stem cells, or hematopoietic stem cells, produce all blood components and are formed before birth in embryos. Raquel Espin Palazons team discovered an immune receptor that activates in embryos, preparing endothelial cells to become stem cells. This finding holds the potential for understanding and manipulating the creation of blood stem cells.

Raquel Espin Palazon said, We know blood stem cells form from endothelial cells, but the factors that set up the cell to switch identity were enigmatic. We didnt know that this receptor was needed or that it was needed this early before blood stem cells even form.

Researchers identified Nod1s role in blood stem cell creation by studying human embryos and using zebrafish. Nod1 levels are closely correlated with blood stem cell development. They collaborated with the Childrens Hospital of Philadelphia to validate this in humans, using induced pluripotent stem cells. Removing Nod1 hindered blood production, confirming its crucial role, similar to its impact on zebrafish blood stem cells.

Researchers, led by Raquel Espin Palazon, found that Nod1 is crucial for blood stem cell development. This discovery opens possibilities for creating blood stem cells from patients samples, a potential game-changer for treating blood disorders without needing bone marrow transplants.

The self-derived stem cells could mitigate risks like graft-versus-host disease. The ongoing research aims to understand the intricate timeline of blood stem cell formation, focusing on developing precise methods. Collaborating with the Childrens Hospital of Philadelphia enhances this effort.

The ultimate goal is therapeutic-grade blood stem cells for curing blood disorder patients. The study involves various Iowa State researchers and collaborators from the University of Edinburgh and Childrens Hospital of Philadelphia.

ISU researchers found a vital step in making blood stem cells. This discovery could create therapeutic-grade stem cells for treating blood disorders, offering a potential breakthrough in regenerative medicine.

The ongoing study focuses on refining methods and understanding the precise timeline of blood stem cell formation. Collaboration with the Childrens Hospital of Philadelphia enhances their efforts. The goal is to provide patients with a revolutionary option, using stem cells derived from their bodies, reducing risks associated with traditional treatments.

Journal reference:

Original post:
Crucial blood stem cell creation step found by ISU researchers - Tech Explorist