Two keys needed to crack three locks for better engineered blood vessels – Penn State University

UNIVERSITY PARK, Pa. Blood vessels engineered from stem cells could help solve several research and clinical problems, from potentially providing a more comprehensive platform to screen if drug candidates can cross from the blood stream into the brain to developing lab-grown vascular tissue to support heart transplants, according to Penn State researchers. Led by Xiaojun Lance Lian, associate professor of biomedical engineering and of biology, the team discovered the specific molecular signals that can efficiently mature nascent stem cells into the endothelial cells that comprise the vessels and regulate exchanges to and from the blood stream.

They published their findings today (March 21) in Stem Cell Reports. The team already holds a patent on foundational method developed 10 years ago and has filed a provisional application for the expanded technology described in this paper.

The researchers found they could achieve up to a 92% endothelial cell conversion rate by applying two proteins SOX17 and FGF2 to human pluripotent stem cells. This type of stem cell, which the researchers derived from a federally approved stem cell line, can differentiate into almost any other cell type if provided the right proteins or other biochemical signals. SOX17 and FGF2 engage three markers in stem cells, triggering a growth cascade that not only converts them to endothelial cells but also enables them to form tubular-like vessels in a dish.

The more efficient differentiation and lab-grown vessels could allow researchers to grow an artificial blood brain barrier to test neurological drugs under development, according to Lian. Other eventual clinical uses may include reestablishing vascular structures after heart damage.

Drugs designed to treat brain diseases need to pass through the blood brain barrier to be effective, Lian said. The blood brain barrier is a membrane packed with vessels and regulates what can pass from the blood into the brain. Our cells can form a tight layer in a dish, onto which we could add various chemicals and see how they pass through.

Next, Lian said, the team will collaborate with industry partners to advance the artificial blood brain barrier and begin testing various drugs. Getting to this point, however, required a decade of investigating the molecular mechanism underpinning how stem cells convert to endothelial cells.

In 2014, we published a protocol using a small molecule that could help the cells differentiate about 20% of the time, but weve now found that just one gene, SOX17, is sufficient for differentiating the about 80% of cells into endothelial cells, said Lian, associate professor of biomedical engineering and of biology at Penn State. That was completely unknown.

In their prior stem cell differentiation process, the low efficiency resulted in heterogenous cell populations, making them difficult to sort and to obtain enough for other research or clinical applications. Lian explained that the researchers knew some of the cells were endothelial cells, but they couldnt predict the other cell types.

To make more homogenous populations, the researchers examined the proteins at play during the process. They first noticed that cells expressed SOX17 during differentiation, so they removed the cells ability to express the protein and analyzed how its absence changed function.

Before knocking down SOX17 expression, about 20% of stem cells would become endothelial cells, Lian said After, differentiation dropped to about 5% at best. We found that SOX17 is required for this process. It was a lucky and surprising finding.

With the addition of SOX17, 80% of stem cells could differentiate. But the researchers wanted to do better, Lian said. The stem cells produce three markers, but SOX17 only triggers two of them to begin the differentiation process. The third marker, called CD31, doesnt work when only exposed to SOX17.

That was a problem for us. We spent two to three years figuring out why, Lian said, explaining that another protein, called FGF2 could induce the marker without affecting SOX17s influence on the other two markers. The combination results in up to 92% of the stem cells differentiating into endothelial cells a more than 350% increase in efficiency from the researchers original approach. Sometimes science is very difficult, but we do not give up.

With all three markers activated, the differentiated cells can form tubular-like vessels in a dish. They can also uptake proteins, like blood vessels in the body. The researchers tested this ability by inducing inflammation to see if the endothelial cells could detect the protein signal involved they could.

Our cells are indeed functional, Lian said. With SOX17 and FGF2, we can determine the fate of these stem cells to be precisely what we need.

Lian is also affiliated with the Materials Research Institute and the Huck Institutes of the Life Sciences at Penn State. Other collaborators on the study include Michael W. Ream, who is a graduate student in the Lian lab in the Department of Biomedical Engineering; Lauren N. Randolph, who earned her doctorate degree in biomedical engineering at Penn State and is now with the San Raffaele Telethon Institute for Gene Therapy in Italy; Yuqian Jian, who also earned her doctoral degree in biomedical engineering at Penn State and is now with the Departments of Pediatrics and of Genetics at Stanford University; and Yun Chang and Xiaoping Bao, both with Purdue Universitys Davidson School of Chemical Engineering.

The U.S. National Science Foundation and the National Institutes of Health funded this research.

More:
Two keys needed to crack three locks for better engineered blood vessels - Penn State University

Stem Cell Editing Repairs Severe Immunodeficiency – The Scientist

The B and T cells of the adaptive immune system recognize unique features on infectious microbes that enter the body. They accomplish this feat using B-cell and T-cell receptors, which take on various shapes to bind to different antigens on foreign invaders. Recombination activating gene 1 (RAG1) is central to this shapeshifting behavior.1 It shuffles the order of DNA sequences in the genes for these receptors, producing multiple versions of the immune receptors that can bind staggering combinations of antigens. However, some people carry mutations in RAG1 that prevent the enzyme from recombining the DNA sequences that code for these receptors. Without properly functioning receptors, B and T cells fail to develop, leading to severe combined immunodeficiency (SCID), a condition in which even the mildest of infections can prove lethal. In a study published in Science Translational Medicine, researchers developed an efficient method to repair RAG1 genes in immune cell progenitors called hematopoietic stem cells (HSC) taken from SCID patients, and revealed that they could restore immune function in mice.2

When you would like to correct the gene, you have to keep in mind that close to the gene, there are a lot of regulatory elements that are relevant for correct gene expression. -Maria Carmina Castiello, San Raffaele Scientific Institute

Maria Carmina Castiello and Anna Villa, two translational immunologists at the San Raffaele Scientific Institute, set out to overcome some of the challenges with editing the RAG1 gene that researchers previously faced. In the past, scientists have taken healthy, functional HSC and inserted them into SCID-model mice, but they often get destroyed by other types of immune cells that recognize the transplants as foreign.3 Normally, doctors use immunosuppressants like chemotherapy before transplantation to deplete immune cells, but this isnt an option for SCID patients. This disease can be associated with severe organ damage, so the critical conditions of the patients do not allow them to receive chemotherapy, Villa explained.

Castiello and her colleagues took a different approach, modifying a SCID patients own stem cells to express a functional RAG1 gene. While other research groups had successfully added RAG1 to patient HSC, they were unable to properly regulate expression of the gene, and therefore couldnt ensure that the stem cells were safe or would effectively replenish B and T cells.

Introducing the gene into the wrong site in the genome may have partly caused this shortcoming. When you would like to correct the gene, you have to keep in mind that close to the gene, there are a lot of regulatory elements that are relevant for correct gene expression, Castiello said.

Rather than adding a functional copy of RAG1, the researchers decided to modify the existing copy, ensuring that the regulatory networks remained intact. In fact, other researchers succeeded when they took a similar approach to edit RAG2.4

Before Castiello and her team could fix the gene, however, they had to choose their editing strategy. Some researchers use base editing, which modifies single letters in the DNA sequence to correct other genetic disorders of these stem cells, like sickle cell disease and -thalassemia.5

Continue reading below...

However, RAG-1mutations can occur at several different sites within the gene, so base editing wouldnt cover every type of mutation. Instead, the research team used the clustered regularly interspaced short palindromic repeats (CRISPR-Cas9) system to cut out a large section of the mutant gene, and then provided cells with the correct DNA sequence using a lentiviral delivery system. Since the correct sequence was nearly identical to the original gene, the cell could swap the sequences unassisted using homology-directed repair (HDR), a built-in DNA repair pathway that fixes double-strand DNA breaks using complementary DNA as a template.

Once Castiello and her colleagues swapped the HSCs old, mutated coding sequence for a fresh one, they had to test whether the gene produced a functional RAG1 protein. They inserted a backwards green fluorescent protein(gfp) gene flanked by sequences that RAG1 recognizes. Promisingly, they found that the edited RAG1 inverted gfp comparably to RAG1 in HSC from healthy donors, thereby switching it to an on state, resulting in a functional gfp gene.

They next had to check that these edited cells could restore immune function in the body. They transplanted these edited human cells into SCID-model mice and found that B and T cells spiked to levels similar to those seen in mice that received HSC from healthy donors.

Whats intriguing from the study is that we dont need to correct all the stem cells. If we manage to correct at least 10 percent of the stem cells, this is going to give us a therapeutic benefit, said Saravanabhavan Thangavel, a geneticist at the Institute of Stem Cell Research and Regenerative Medicine who was not involved with the work. However, he also mentioned, We need to track the HDR-edited cells long term. The researchers need to ensure that the modified cells persist in the bodies of people with SCID so that their newly gained immunity doesnt wane over time. If, by chance, the HDR-edited cells faded away, they may not have a therapeutic benefit, Thangavel added.

Down the line, the team aims to refine their protocol. We are trying to increase the editing efficiency that we achieve, Castiello said. She also wants to optimize delivery of the gene into the cells by comparing different methods. In this study they used lentiviruses to deliver the DNA template to the stem cells, but they plan to test other strategies like using lipid nanoparticle conduits that conceal the DNA template and fuse with the cell membrane to release the DNA into the cell.

The team will also have to test the safety of this gene editing strategy and find a way to scale up production of the edited stem cells, Castiello added. Then they should be able to test their edited cells in people with the hope of eventually treating the variety of conditions caused by RAG1defects. We are really committed to translating our strategy to the clinic, she said.

Continue reading below...

Anna Villa and Maria Carmina Castiello are inventors with two patents involved with editing RAG genes.

The rest is here:
Stem Cell Editing Repairs Severe Immunodeficiency - The Scientist

Discovery could revolutionize stem cell-based brain repair therapy for Parkinson’s disease – News-Medical.Net

Neuroscientists at University of Galway have made an exciting discovery that could revolutionize stem cell-based brain repair therapy for Parkinson's disease.

Parkinson's is a neurodegenerative condition in which brain cells slowly degenerate and die leading to a progressive deterioration in a person's ability to control movement. It is estimated that there are 8.5 million people living with the condition worldwide, and 12,000 people in Ireland alone.

Brain repair for Parkinson's involves replacing the dead cells by transplanting healthy brain cells back into the brain. With recent advancements in regenerative medicine and stem cell technology, "induced stem cells" can now be used as a source of healthy cells.

Induced stem cells are reprogrammed from adult cells, such as skin cells, and can be converted in the laboratory into the appropriate type of brain cell required for repairing the Parkinson's brain.

However, these skin cells-turned brain cells need to be transplanted into the brain at a very early stage in their conversion, and the vast majority of the cells do not continue to convert -once in the brain - into the mature cells that are required for the therapy to work.

In work funded by The Michael J. Fox Foundation for Parkinson's Research (MJFF) and Science Foundation Ireland, published this week in the Journal of Neural Engineering, the team in the College of Medicine, Nursing and Health Sciences at University of Galway have shown that transplanting the immature cells in a collagen hydrogel dramatically improves both their survival and maturation in the brain.

Commenting on the research finding, the lead neuroscientist on the project, Professor Eils Dowd, said:"Our hydrogel nurtures, supports and protects the cells after they are transplanted into the brain, and this dramatically improves their maturation and reparative ability. Ultimately, we hope that continued development of this promising gel will lead to a significant improvement in brain repair approaches for people living with Parkinson's."

The Michael J. Fox Foundation awarded $300,000 to continue the development of the hydrogel.

The new research aims to understand how the immune system in the brain reacts when cells are transplanted alone versus when they are transplanted in combination with the hydrogel.

The research will continue to be led by Professor Dowd, in collaboration with colleagues from CRAM - the Science Foundation Ireland Research Centre for Medical Devices based at University of Galway, the University of Edinburgh and the University of Melbourne.

Professor Dowd's ongoing research in this field is featured in the short documentary Feats of Modest Valourwhich won the coveted Scientist Award at the Imagine Science Film Festival in New York, as well as the Professional Documentary Award at the Raw Science Festival in California.

Source:

Journal reference:

Comini, G.,et al.(2024) Survival and maturation of human induced pluripotent stem cell-derived dopaminergic progenitors in the Parkinsonian rat brain is enhanced by transplantation in a neurotrophin-enriched hydrogel.Journal of Neural Engineering.doi.org/10.1088/1741-2552/ad33b2.

Here is the original post:
Discovery could revolutionize stem cell-based brain repair therapy for Parkinson's disease - News-Medical.Net

Collagen hydrogel can boost survival of precursor neurons to brain – Parkinson’s News Today

Encapsulating precursor nerve cells in a collagen hydrogel can enhance the efficacy of stem cell transplantation to the brain, a potential treatment to replace dopamine-producing nerve cells that are lost in people with Parkinsons disease, according to a preclinical study.

Our hydrogel nurtures, supports and protects the cells after they are transplanted into the brain, and this dramatically improves their maturation and reparative ability, Eils Dowd, PhD, study lead at the College of Medicine, Nursing and Health Sciences at the University of Galway, in Ireland, said in a university press release.

Ultimately, we hope that continued development of this promising gel will lead to a significant improvement in brain repair approaches for people living with Parkinsons, Dowd said.

The study, Survival and maturation of human induced pluripotent stem cell-derived dopaminergic progenitors in the Parkinsonian rat brain is enhanced by transplantation in a neurotrophin-enriched hydrogel, supported by research grants from The Michael J. Fox Foundation for Parkinsons Research (MJFF), was published in the Journal of Neural Engineering.

Parkinsons symptoms are caused by the progressive loss of dopaminergic neurons, the specialized nerve cells that produce dopamine, a chemical messenger in the brain.

Cell-based brain repair is a promising therapeutic approach for Parkinsons disease, whereby the lost dopaminergic neurons are replaced by the transplantation of healthy neurons. Such neurons can be derived from induced pluripotent stem cells, or iPSCs, that are reprogrammed from adult cells, such as skin cells, and converted into dopaminergic neurons.

In fact, stem cell-based therapies have already started clinical testing, including a trial evaluating bemdaneprocel, which demonstrated sustained efficacy 1.5 years after treatment, and another called STEM-PD.

However, iPSCs need to be transplanted into the brain very early in their maturation from stem cells to fully functional neurons. Even so, most transplanted cells do not fully mature into dopaminergic neurons once they are inside the brain.

Thus, it is imperative to continue rigorous preclinical studies to identify methods to improve their outcome to maximise their reparative/reconstructive potential, Dowd and her colleagues wrote.

Encapsulating cells in hydrogels made with collagen, the most abundant protein in connective tissue, has the potential to address many limitations of transplantation. These hydrogels act as a scaffold, shielding cells from immune responses, and can be infused with neurotrophic factors, proteins that induce the survival, development, and function of neurons.

Ultimately, we hope that continued development of this promising gel will lead to a significant improvement in brain repair approaches for people living with Parkinsons.

In previous work, Dowds team encapsulated primary dopaminergic neurons in a collagen hydrogel loaded with a neurotrophic factor called GDNF. Transplantation with the specialized gel dramatically increased the survival and function of these cells in a rat model of Parkinsons.

In this new study, the researchers tested the gel method using dopaminergic precursor cells (DAPs) derived from iPSCs (iPSC-DAPs). Cells were transplanted into the brain of a Parkinsons rat model, with or without the gel, and with or without the neurotrophic factors GDNF and BDNF.

Surviving iPSC-derived cells were visible in all four groups as early as one week after transplantation. Still, the graft cells were significantly larger in the two groups treated with iPSC-DAPs in the hydrogel, with or without neurotrophic factors, than in the two groups without the hydrogel.

This suggested not only was the hydrogel cytocompatible with these cells, the beneficial effects of the gel were already manifesting shortly after transplantation, the researchers wrote.

Although the cells transplanted with the hydrogel stimulated a broader immune response, the researchers noted this was likely a reflection of the larger graft size in the hydrogel groups.

As assessed 20 weeks, or 4.6 months, after transplantation, neurotrophic-enriched iPSC-DAPs in the hydrogel were larger and more dispersed within the brain compared with cells transplanted alone, which were small and compact. When transplanted with the neurotrophic-enriched hydrogel, cell survival increased eightfold, and dopaminergic neuron maturation improved 16 times higher.

Comparably, significantly more iPSC-DAPs matured into dopaminergic neurons with the enriched hydrogel than without (11.2% vs. 2.1%). Furthermore, one in six rats given cells alone reached a dopaminergic maturation level of more than 5%, whereas all seven rats treated with enriched cells in the hydrogel achieved this outcome.

This suggests that the neurotrophin-enriched hydrogel is having a beneficial effect on dopaminergic differentiation and maturation over and above the effect on progenitor survival, the team wrote.

Given the beneficial effects of this hydrogel on human iPSC-derived brain repair in this Parkinsonian rat model, further development of such hydrogel carriers is warranted to improve the survival, differentiation and overall outcome of stem cell-derived brain repair in Parkinsons patients, the researchers concluded.

Further development of the hydrogel, specifically to understand how the immune system in the brain reacts upon transplantation, is being supported by a $300,000 award from the MJFF.

See the original post here:
Collagen hydrogel can boost survival of precursor neurons to brain - Parkinson's News Today

Scientists Uncover Cause of Inherited Bone Marrow Failure – Mirage News

An international study led by researchers from Children's Hospital of Philadelphia (CHOP) has discovered an important biological cause of Fanconi anemia, a rare inherited disorder that almost universally leads to bone marrow failure. The researchers also confirmed that a readily available bile acid may help correct some of these biological issues and provide more options for potential treatment. The findings were recently published by the journal Nature Communications.

Fanconi anemia was discovered nearly a century ago. Over the course of decades of study, researchers had previously linked Fanconi anemia to DNA damage that impairs the function of hematopoietic stem cells (HSCs), which are responsible for lifelong function and renewal of blood cells. DNA damage leads to cell death and eventually gives way to frank bone marrow failure. Since stem cells are impacted, a stem cell transplant is the only treatment currently available for Fanconi anemia. However, not all patients have matching donors, and those who do can still experience side effects from the treatment.

Prior research by the group revealed that the loss of stem cells that leads to Fanconi anemia begin before birth, making the underlying biology behind this condition very difficult to study. Using mouse models and a combination of other techniques, researchers were able to determine the events before birth that eventually accelerate bone marrow failure by early school age in these patients.

Peter Kurre, MD "Fanconi anemia is thought to be a DNA disorder, and those who have been researching it have focused solely on this aspect," said senior author Peter Kurre, MD, Director of the Pediatric Comprehensive Bone Marrow Failure Center at CHOP. "What our research demonstrates for the first time is that Fanconi anemia is caused by an accumulation of misfolded proteins that preclude proper cell cycle progression and stem cell expansion, which in turn leads to rapid bone marrow failure after birth."

The study found that increased protein synthesis rates in fetal HSCs occur at the onset of Fanconi anemia. The proteins are misfolded in fetal liver HSCs, which leads to stress on the endoplasmic reticulum, a network of tubules in our cells responsible for producing healthy proteins needed for a variety of functions. This accumulation of misfolded proteins confers cellular stress signals that slow down cell cycle progression and emergence of a sufficient number of stem cells, causing rapid bone marrow failure early in life.

With this new knowledge of the mechanisms behind Fanconi anemia, the researchers then used tauroursodeoxycholic acid (TUDCA), a bile salt that has been studied for use in preventing and treating gallstones and helping to reduce the side effects associated with certain inflammatory disorders. In HSC of the Fanconi anemia animal model, TUDCA restored proper protein folding.

"When TDUCA was given, we were able to confirm the restoration of folding as well as improved function of the stem cells," said Narasaiah Kovuru, PhD, a postdoctoral researcher in the Kurre Laboratory and first author of the study.

With this combination of information, the researchers determined that the degradation of protein homeostasis - the proper regulation of proteins in these cells - is driven by excess inflammatory activity in HSCS within the fetal liver, and dampening this activity helps restore HSCs to their normal numbers.

"Regulating protein production, folding and degradation properly is like the story of Goldilocks and the Three Bears, in that too much or too little protein is a problem, and the levels need to be just right for proper function," Kurre said. "This study reveals the direction our research should go next to determine what mechanisms are at play that alter these protein levels and what alternate treatment options we might be able to develop for patients for whom stem cell transplantation is not an option."

This study was supported by National Institutes of Health grant R01-HL150882.

Kovuru et al, "Deregulated protein homeostasis constrains fetal hematopoietic stem cell pool expansion in Fanconi anemia." Nat Commun. Online February 29, 2024. DOI: 0.1038/s41467-024-46159-1.

See the original post:
Scientists Uncover Cause of Inherited Bone Marrow Failure - Mirage News

CuSTOM Organoid Research Evolving From Tool to Treatment – Research Horizons – Research Horizons

Two scientists at Cincinnati Childrens describe the fast-evolving field of organoid medicine as todays research tool, tomorrows organ transplant solution.

A special feature published March 14, 2024, in Science explores advances made by the Cincinnati Childrens Center for Stem Cell and Organoid Medicine (CuSTOM). The article features organoid experts Takanori Takebe, MD, PhD, and Mingxia Gu, MD, PhD.

These 3D formations, grown from induced pluripotent stem cells (iPSCs), are living mini-organs that include the key cell types of full-sized organs.

Already, organoids are showing value as test platforms for preclinical drug testing and ongoing basic studies of human development. Longer term, experts at CuSTOM envision organoids grown from a patients own cells serving as a new method to treat disease and repair organ damage.

Read more about CuSTOM in Science.

Continued here:
CuSTOM Organoid Research Evolving From Tool to Treatment - Research Horizons - Research Horizons

UCF Researchers Develop Novel Therapy for Incurable Brain Cancer – UCF

College of Medicine researchers are developing a more effective way to treat glioblastoma an aggressive, incurable form of brain cancer. Patients currently live just 12 to 15 months after diagnosis despite surgery, radiation and chemotherapy.

New research led by Kiminobu Sugaya, a stem cell researcher and neuroscientist at UCFs Burnett School of Biomedical Sciences, found that targeting a drug resistant mechanism in cancer stem cells significantly enhanced the efficacy of traditional cancer therapies making them four times more effective against glioblastoma. Current FDA-approved drugs kill less than 25% of glioblastoma cancer stem cells (CSCs).

These cells are a subpopulation of cancer cells that are highly resistant to current therapies. Scientists theorize that cancer returns and spreads because CSCs remain in the body. Thats why they are exploring ways to kill them outright.

Cancer stem cells are bad stem cells that are programed to become a cancer, Sugaya says. They withstand cancer therapies, raise their ugly head, regrow and metastasize.

Sugayas team developed a new drug delivery system by creating a technology that destroys the RNA, or ribonucleic acid, that the stem cells use as a blueprint to produce proteins. This unique strategy inhibits the expression of embryonic stem cell genes that are pivotal in CSCs drug resistance. And because embryonic stem cell genes are not expressed in normal adult cells, this breakthrough approach reduces potential for side effects in healthy cells.

Jonhoi Smith is a doctoral student under Sugaya and the first author on their research paper published in the journal Genes. He said the treatment could increase life expectancy for glioblastoma patients.

This treatment could be a precious gift for glioblastoma patients. When I think about the loved ones Ive lost in my life my father, my grandmother I often wish I could have had more time with them, he says. The idea of offering the potential of a whole new life to people who are facing a death sentence in less than a year means a lot to me.

One of the significant challenges in treating glioblastoma is effectively delivering treatments to the brain. Thats because the brain is protected from external germs and substances by the blood-brain barrier, which can also prevent treatments from reaching brain tissues.

To overcome this obstacle, Sugayas therapy is based on exosomes, nano-sized particles with a lipid membrane that are naturally produced by cells. Exosomes function as cellular communicators, transporting proteins, lipids and genetic material between cells, thereby influencing a wide array of biological processes and functions. Their efficiency in carrying molecules across various parts of the body has inspired scientists to investigate exosomes as potential drug delivery vehicles.

Many current drug delivery systems, including viruses, may cause side effects, Sugaya explains. Were using the bodys natural delivery systems and have developed technologies to modify them to carry therapeutic molecules with targeted delivery to specific tissues.

Marvin Hausman is CEO of Exousia AI, the company that is funding the glioblastoma exosome preclinical research. He heard about Sugayas lab and says that when he visited the lab at UCFs Academic Health Sciences Campus in Lake Nona, he was inspired by its capacity for innovative discoveries.

I have thoroughly analyzed this exosome-based targeted drug delivery system many times, and the potential that this unique technology offers. Hausman says. We are embarking on a revolutionary new development in medicine.

Thanks to funding from Exousia AI, the research is advancing to mouse models carrying human glioblastoma, with preliminary results expected as early as this summer.

Sugaya has dedicated more than 40 years to neuroscience research focused on Alzheimers disease, with an emphasis on stem cells for the last 26 years. He moved to the U.S. after receiving his doctoral degree from the Science University of Tokyo in 1988. He joined UCF as a professor in 2004. His cancer research began in 2010 when he discovered stemness gene expressions, the self-renewing and differentiating property that allows stem cells to grow and spread, in CSCs. He is recognized as an expert in the field of exosome research and recently received Florida Innovation Funding from the State Department of Health for his studies.

The rest is here:
UCF Researchers Develop Novel Therapy for Incurable Brain Cancer - UCF

BioCardia and StemCardia Announce Biotherapeutic Delivery Partnership – Diagnostic and Interventional Cardiology

March 15, 2024 BioCardia, Inc., a biotechnology company focused on advancing late-stage cell therapy interventions for cardiovascular disorders, andStemCardia, Inc., a biotechnology company focused on cell and gene therapy to re-muscularize the failing heart, today announced a long-term partnership to advance StemCardias investigational pluripotent stem cell product candidate for the treatment of heart failure.

Under the partnership, BioCardia is the exclusive biotherapeutic delivery partner for StemCardias cell therapy candidate through studies expected to result in FDA approval of an investigational new drug application (IND) and the anticipated Phase I/II clinical development to follow.

BioCardia has established safe and minimally invasive delivery of cellular medicines directly into the heart, said Chuck Murry, MD, PhD, StemCardias Founder and CEO. Having worked with BioCardia to successfully deliver our bona fide cardiac muscle cells in large animal models of heart failure, we are excited for this partnership to accelerate clinical development and broaden future commercial access to an off-the-shelf heart regeneration treatment.

StemCardias team encompasses recognized leaders in the field of cardiac regenerative medicine who are pursuing an elegant strategy to repair the failing heart. We look forward to supporting their efforts with our experienced team and proven, proprietary Helix biotherapeutic delivery system, said BioCardia CEO Peter Altman, PhD. This partnership is expected to enhance future treatment options for millions of people suffering from heart failure, offset the costs of biotherapeutic delivery development for our own programs, and provide our investors with meaningful revenue sharing should our efforts together contribute to StemCardias successful therapeutic development.

For more information:www.biocardia.com

See the rest here:
BioCardia and StemCardia Announce Biotherapeutic Delivery Partnership - Diagnostic and Interventional Cardiology

New Positive Data Presented on Briquilimab Conditioning in Patients with Fanconi Anemia – GlobeNewswire

REDWOOD CITY, Calif., March 15, 2024 (GLOBE NEWSWIRE) -- Jasper Therapeutics, Inc. (Nasdaq: JSPR) (Jasper), a biotechnology company focused on development of briquilimab, a novel antibody therapy targeting c-Kit (CD117) to address mast cell driven diseases such as chronic spontaneous urticaria (CSU) and chronic inducible urticaria (CIndU), announced additional positive Phase 1b/2a data on briquilimab as a conditioning agent in the treatment of Fanconi Anemia (FA).

The data was presented at the 2024 Stanford Medicine Center for Definitive and Curative Medicine Symposium on March 13, 2024, in Palo Alto, California.

The ongoing investigator initiated Phase 1b/2a clinical trial is evaluating a conditioning regimen that includes intravenous briquilimab as a potential treatment for FA patients in bone marrow failure. Data from the study show that briquilimab infusion has a promising safety profile and appears to be well tolerated in patients with FA, with all six patients treated achieving full donor engraftment and full blood count recovery.

We continue to be encouraged by the results from Stanford Medicine's Phase 1b/2a study in Fanconi Anemia, which demonstrates the potential of briquilimab to serve as a key component of non-toxic conditioning regimens for stem cell transplant, said Edwin Tucker, Chief Medical Officer of Jasper. Wed like thank our collaborators at Stanford Medicine for their work evaluating briquilimab in this vulnerable patient population.

About Briquilimab

Briquilimab (formerly JSP191) is a targeted aglycosylated monoclonal antibody that blocks stem cell factor from binding to the cell-surface receptor c-Kit, also known as CD117, thereby inhibiting signaling through the receptor. This inhibition disrupts the critical survival signal, leading to the depletion of the mast cells via apoptosis which removes the underlying source of the inflammatory response in mast cell driven diseases such as chronic urticaria. Jasper is currently conducting clinical studies of briquilimab as a treatment in patients with CSU or with CIndU. Briquilimab is also currently in clinical studies as a treatment for patients with LR-MDS and as a conditioning agent for cell and gene therapies for rare diseases. To date, briquilimab has a demonstrated efficacy and safety profile in more than 145 dosed participants and healthy volunteers, with clinical outcomes as a conditioning agent in severe combined immunodeficiency (SCID), acute myeloid leukemia (AML), myelodysplastic syndromes (MDS), FA, and sickle cell disease (SCD).

About Fanconi Anemia

Fanconi Anemia (FA) is a rare but serious blood disorder that prevents the bone marrow from making sufficient new red blood cells. The disorder can also cause the bone marrow to make abnormal blood cells. FA typically presents at birth or early in childhood between five and ten years of age. Ultimately, it can lead to serious complications, including bone marrow failure and severe aplastic anemia. Cancers such as acute AML and MDS are other possible complications. Treatment may include blood transfusions or medicine to create more red blood cells, but a hematopoietic stem cell transplant (HSCT) is currently the only cure.

About Phase 1/2 clinical trial (NCT04784052)

The Stanford sponsored, investigator initiated Phase 1/2 study is an open-label clinical trial evaluating briquilimab as a potential treatment for FA patients in bone marrow failure (BMF) requiring allogeneic transplant. Utilizing briquilimab, the regimen eliminates the need for busulfan chemotherapy or total body irradiation. Participants with FA with BMF receive allo-HCT with TCR+ T-cell/CD19+ B-cell depleted hematopoietic grafts from 10/10 unrelated, 9/10 unrelated or haploidentical family donors. A 0.6 mg/kg dose of briquilimab is administered in combination with standard FA dosing of anti-thymocyte globulin (ATG), cyclophosphamide, fludarabine, and rituximab as lymphodepletion. The primary outcomes include safety, efficacy, and engraftment success.

About Jasper

Jasper is a clinical-stage biotechnology company developing briquilimab, a monoclonal antibody targeting c-Kit (CD117) as a therapeutic for chronic mast and stem cell diseases such as chronic urticaria and lower to intermediate risk MDS and as a conditioning agent for stem cell transplants for rare diseases such as SCD, FA and SCID. To date, briquilimab has a demonstrated efficacy and safety profile in more than 145 dosed participants and healthy volunteers, with clinical outcomes as a conditioning agent in SCID, acute myeloid leukemia, MDS, FA, and SCD. For more information, please visit us atwww.jaspertherapeutics.com.

Forward-Looking Statements

Certain statements included in this press release that are not historical facts are forward-looking statements for purposes of the safe harbor provisions under the United States Private Securities Litigation Reform Act of 1995. Forward-looking statements are sometimes accompanied by words such as believe, may, will, estimate, continue, anticipate, intend, expect, should, would, plan, predict, potential, seem, seek, future, outlook and similar expressions that predict or indicate future events or trends or that are not statements of historical matters. These forward-looking statements include, but are not limited to, statements regarding briquilimabs potential, including its potential as a conditioning agent in the treatment of FA and FA patients in bone marrow failure and its safety profile, its potential to serve as a key component of non-toxic conditioning regimens for stem cell transplant and its potential to address mast cell driven diseases such as CSU and CIndU. These statements are based on various assumptions, whether or not identified in this press release, and on the current expectations of Jasper and are not predictions of actual performance. These forward-looking statements are provided for illustrative purposes only and are not intended to serve as, and must not be relied on by an investor as, a guarantee, an assurance, a prediction or a definitive statement of fact or probability. Many actual events and circumstances are beyond the control of Jasper. These forward-looking statements are subject to a number of risks and uncertainties, including general economic, political and business conditions; the risk that the potential product candidates that Jasper develops may not progress through clinical development or receive required regulatory approvals within expected timelines or at all; the risk that clinical trials may not confirm any safety, potency or other product characteristics described or assumed in this press release; the risk that Jasper will be unable to successfully market or gain market acceptance of its product candidates; the risk that prior study results may not be replicated; the risk that Jaspers product candidates may not be beneficial to patients or successfully commercialized; patients willingness to try new therapies and the willingness of physicians to prescribe these therapies; the effects of competition on Jaspers business; the risk that third parties on which Jasper depends for laboratory, clinical development, manufacturing and other critical services will fail to perform satisfactorily; the risk that Jaspers business, operations, clinical development plans and timelines, and supply chain could be adversely affected by the effects of health epidemics; the risk that Jasper will be unable to obtain and maintain sufficient intellectual property protection for its investigational products or will infringe the intellectual property protection of others; and other risks and uncertainties indicated from time to time in Jaspers filings with the SEC, including its Annual Report on Form 10-K for the year ended December 31, 2023 and subsequent Quarterly Reports on Form 10-Q. If any of these risks materialize or Jaspers assumptions prove incorrect, actual results could differ materially from the results implied by these forward-looking statements. While Jasper may elect to update these forward-looking statements at some point in the future, Jasper specifically disclaims any obligation to do so. These forward-looking statements should not be relied upon as representing Jaspers assessments of any date subsequent to the date of this press release. Accordingly, undue reliance should not be placed upon the forward-looking statements.

Contacts:

Joyce Allaire (investors) LifeSci Advisors 617-435-6602 jallaire@lifesciadvisors.com

Alex Gray (investors) Jasper Therapeutics 650-549-1454 agray@jaspertherapeutics.com

Lauren Walker (media) Real Chemistry 646-564-2156 lbarbiero@realchemistry.com

See the rest here:
New Positive Data Presented on Briquilimab Conditioning in Patients with Fanconi Anemia - GlobeNewswire

Notoginsenoside R1 promotes Lgr5+ stem cell and epithelium renovation in colitis mice via activating Wnt/-Catenin … – Nature.com

Chemicals and reagents

Notoginsenoside R1 (NGR1, BP1010, C47H80O18, purity 98%, CAS No 80418-24-2, MW: 933.13Da) was purchased from Chengdu Purifa Technology Development Co. Ltd (Chengdu, China). Dextran sulfate sodium salt (DSS, 0216011010, MW: 36kDa50kDa) was purchased from MP Biomedicals (Shanghai, China). Salicylazosulfapyridine (SASP, S0883, C18H14N4O5S, CAS No 599-79-1, MW: 398.39Da) and FITC-dextran (FD4, CAS No 60842-46-8) was purchased from Sigma-Aldrich (Darmstadt, Germany). ICG-001 (T6113, C33H32N4O4, purity 98%, CAS No 780757-88-2, MW: 548.64Da) was acquired from TOPSCIENCE (Shanghai, China). Water-DEPC treated (693520) and DMSO (D8418) were obtained from MilliporeSigma (Burlington, MA, USA).

NCM460 human intestinal epithelial cells and CT26 murine colon carcinoma cells were obtained from the American Type Culture Collection (ATCC, Manassas, VA, USA). NCM460 and CT26 cells were cultured in Roswell Park Memorial Institute (RPMI)-1640 culture medium (11875085, Gibco, NY, USA) supplemented with 10% fetal bovine serum (10099158, Gibco, NY, USA). The culture conditions included a humidified atmosphere containing 5% CO2, with a constant temperature maintained at 37C.

The Laboratory Animal Center of Shanghai University of Traditional Chinese Medicine provided female C57BL/6 mice weighing 202g. These mice were housed in a specific pathogen-free facility under meticulously controlled conditions, including a temperature range of 2325C, humidity maintained at 60%70%, and a well-regulated 12-h light-dark cycle. The Animal Experimentation Ethics Committee of Shanghai University of Traditional Chinese Medicine granted approval (PZSHUTCM2307310004) for experimental procedures conducted on the animals. All experiments were conducted in accordance with institutional animal care guidelines and protocols approved by the committee.

According to the method reported by Yue [26], we established the acute colitis mouse model. Briefly, female C57BL/6 mice were divided randomly into four groups: Control, DSS, DSS+SASP, and DSS+NGR1. Acute colitis was induced by administering 3% DSS in the drinking water of mice for a period of 8 days. Mice in the DSS+SASP group were treated orally with SASP (260mg/kg) once per day for the same duration. The DSS+NGR1 group received NGR1 (25, 50, 125mg/kg) by oral gavage once per day for 10 days. Mice in the Control and DSS groups were administered the same volume of Control. Daily monitoring of body weight and rectal bleeding was conducted throughout the 10-day period. At the end of the experiment, mice were euthanized, and the colon was collected for further analysis.

Female C57BL/6 mice were randomly divided into four groups: DSS, DSS+ICG-001, DSS+NGR1 and DSS+ICG-001+NGR1. To establish an acute enteritis model, mice were subjected to the protocol described above. Mice in the DSS+NGR1 and DSS+ICG-001+NGR1 group were given NGR1 (25mg/kg) orally once daily for 10 consecutive days. Meanwhile, mice in the DSS+ICG-001 and DSS+ICG-001+NGR1 groups were given ICG-001 (20mg/kg) via intraperitoneal injection three times per week. The DSS and DSS+NGR1 groups received the same volume of Control.

Male BALB/c mice were acclimated for 1 week in a specific pathogen-free environment. Subsequently, CT26 cells (2105 cells/mouse) were subcutaneously transplanted into the left axillary region of each mouse. Once the tumor size reached 200mm3, the mice were randomly assigned to the vehicle group or the NGR1 group based on tumor size. Throughout the 18-day experiment, mice in the vehicle group received 0.5% CMC-Na, while those in the NGR1 group were administered 25mg/kg NGR1. Tumor volume=0.5length (mm)width (mm)2.

C57BL/6 mice were fasted for 4h before execution. Mice were then orally administered 60mg/100g body weight of FITC-dextran in 200L of sterile saline. After 4h, blood samples were collected via retro-orbital bleeding, and serum was separated by centrifugation. The serum FITC-dextran levels were measured at an excitation wavelength of 485nm and an emission wavelength of 528nm using a fluorometer (VARIOSKAN FLASH, Thermo Fisher, MA, USA).

Colonic tissues were collected from mice and fixed in 4% paraformaldehyde. Tissues were then dehydrated, embedded in paraffin, and sectioned into 4m thick slices. The sections were then stained with hematoxylin and eosin (H&E) using standard protocols. Stained sections were analyzed under a light microscope (BX61VS, Olympus, Tokyo, Japan), and images were captured for further analysis.

The concentrations of DAO (CSB-E10090m) and LPS (CSB-E13066m) in mouse serum samples were determined using the respective ELISA kit (Wuhan Huamei Biological Engineering Co., Ltd, Wuhan, China). Specifically, serum samples were added to a 96-well plate coated with DAO or LPS-specific antibodies, followed by incubation with detection reagents and substrate solution. Absorbance was measured at 450nm, and concentrations were calculated using standard curves.

Colonic tissues were fixed in 4% paraformaldehyde, embedded in OCT compound, and sectioned into 5-m slices. After permeabilization and blocking, sections were incubated with primary antibodies against ZO-1 (#13663, Cell Signaling Technology, CST, MA, USA) and Occludin (#91131, CST, MA, USA), followed by secondary antibodies conjugated to fluorophores (9300039001, ABclonal, Wuhan, China). Nuclei were counterstained with DAPI (#4083, CST, MA, USA), and images were obtained using a fluorescence microscope (BX61VS, Olympus, Tokyo, Japan). Quantification of ZO-1 and Occludin expression was performed using ImageJ software (NIH, Bethesda, MD, USA).

Colonic tissue samples were obtained from mice, fixed, dehydrated, embedded in paraffin blocks, sectioned, and stained with Alcian blue using a commercial kit. Under a light microscope (BX61VS, Olympus, Tokyo, Japan), the stained sections were examined and images were captured for subsequent analysis.

RNA was extracted using the TRIzol method, and RNA quantity and purity were measured by NanoDrop spectrophotometer (Thermo Fisher Scientific). The RNA was then reverse-transcribed using an Evo M-MLV RT Premix for qPCR kit (AG11706, Accurate Biotechnology Co., Ltd., Chengdu, China), and qPCR was performed using a SYBR Green Premix Pro Taq HS qPCR Kit (AG11718, Accurate Biotechnology Co., Ltd., Chengdu, China) (Table1). The amplification was carried out using an ABI Prism 7900HT Sequence Detection System (Life Technologies, CA, USA), and data were analyzed using the 2Ct method.

Colonic tissues were extracted and homogenized, and protein was obtained using RIPA lysis buffer with phosphatase and protease inhibitors. Protein concentration was measured using a BCA assay kit (20201ES76, Yeasen Biotech Co., Ltd, Shanghai, China). Equal amounts of protein were loaded onto SDS-PAGE (sodium dodecyl sulfate polyacrylamide gel electrophoresis) gels and separated by electrophoresis. Subsequently, the separated proteins were transferred onto PVDF membranes (000025736, Milipore, MA, USA). The membrane was then blocked with 5% BSA solution for 2h. After blocking, the membrane was incubated with primary antibodies overnight at 4C, followed by incubation with HRP-conjugated secondary antibodies for 1h at room temperature. Protein bands were visualized using ECL reagents (WBKLS0500, Millipore) and imaged with a GS-700 imaging densitometer (Bio-Rad, CA, USA). Protein expression levels were quantified using ImageJ software (NIH, Bethesda, MD, USA). The following primary antibodies were used: rabbit anti--Catenin (1:1000, #8480, CST, MA, USA), rabbit anti-p-GSK-3 (1:1000, #5558, CST, MA, USA), rabbit anti-GSK-3 (1:1000, #12456, CST, MA, USA), rabbit anti-Cyclin D1 (1:1000, #2922, CST, MA, USA), rabbit anti-c-Myc (1:1000, #5605, CST, MA, USA) and rabbit anti--actin (1:1000, #4970, CST, MA, USA).

Colonic tissue sections were fixed in 4% paraformaldehyde, embedded in paraffin, and sliced into 5m thick sections. Antigen retrieval was performed using citrate buffer solution (pH=6.0) and heating in a microwave oven. Non-specific binding was blocked with 5% goat serum for 30min. Sections were incubated overnight at 4C with primary antibodies, followed by incubation with a secondary antibody and staining with DAB (3,3-diaminobenzidine). Hematoxylin was used for counterstaining before the sections were examined microscopically and images were captured.

Total RNA was extracted from mouse intestinal tissues using TRIzol reagent according to the manufacturers instructions. The extracted RNA was evaluated for quality using a NanoDrop spectrophotometer (Thermo Fisher). RNA sequencing libraries were then constructed with the NEBNext Ultra RNA Library Prep Kit for Illumina, and sequencing was performed on an Illumina HiSeq platform. The differential gene was carried out on the cloud platform of majorbio (https://www.majorbio.com/).

Caco-2 cells were seeded in Millicell inserts of 24-well plates at a density of 5104 cells/400L per well. The outer chamber was filled with 600L DMEM medium (2323012, Gibco, NY, USA) and replaced every other day. TEER values were measured using a MERS00002 volt-ohm meter system (Milipore), and the electrode was sterilized with 70% ethanol and rinsed with sterile phosphate-buffered saline (PBS) before each measurement. Monolayer formation was assumed at TEER values of 400/cm2. Measurements were taken at regular intervals using the same electrode and recorded.

The intestinal crypts were isolated from the small intestine of C57BL/6 mice (6- to 8-week-old). The small intestine was removed and flushed with ice-cold PBS. The intestine was opened longitudinally and cut into 2- to 3-mm pieces. The pieces were then washed with ice-cold PBS and incubated in 3mM EDTA solution at 4C for 20min with gentle shaking. After incubation, the crypts were released by vigorously shaking the tubes. The supernatant containing the crypts was collected and filtered through a 70-m cell strainer. The crypts were then centrifuged at 1200r/min for 5min and resuspended in Matrigel (Corning, NA, USA). The crypt-Matrigel mixture was plated in 24-well plates and incubated at 37C for 30min to allow the Matrigel to solidify. The IntestCultTM OGM Mouse Basal Medium (#06005, STEMCELL, Vancouver, Canada) was then added to the wells and changed every other day.

After cultured 2 days in a 24 well plate, the intestinal crypts were randomly divided into control, DSS model group and DSS+NGR1 group. Then, the organoids were administered DSS (20g/mL), DSS (20g/mL) plus NGR1 (100M) for 4 days. The organoid growth conditions were recorded by the microscope (Olympus CKX4, Tokyo, Japan). IHC assay was conducted to examine the fluorescent protein expression of Lgr5 and -Catenin (refer to the above method of IHC).

The molecular docking was performed using AutoDock Vina software. The 3D crystal structure of -Catenin protein (PDB: 1JDH) was obtained from the Protein Data Bank (PDB) database. The structure of NGR1 was drawn and optimized using ChemDraw software and converted to a PDB file using Open Babel software. The protein and ligand files were prepared using AutoDock Tools. Docking simulations were performed and the conformation with the lowest binding energy was selected as the final docking result. The docking results were analyzed using PyMOL software.

The TOPFlash assay was performed as previously described with slight modifications [27]. HEK293T cells were seeded in 24-well plates and cultured overnight. The cells were transfected with the 500ng TOPFlash luciferase reporter plasmids (Beyotime Biotechnology, Shanghai, China) and 50ng Renilla luciferase (Promega GmbH, Mannheim, Germany) using Lipofectamine 3000 (Thermo Fisher). After 24h, the cells were treated with NGR1 (50M) and BIO (0.5M) for 24h, separately. Subsequently, cells were lysed in 150L/well passive phenylbenzothiazole (PPBT) buffer, and the luciferase activity was measured using a Dual-LuciferaseTM Reporter Assay System (Promega Corporation, WI, USA). The firefly luciferase activity was normalized to Renilla luciferase activity.

A scratch wound was created using a plastic pipette (10L) tip. NCM460 cells were then washed with PBS to remove any debris and treated with either DSS (20g/mL) or DSS (20g/mL)+NGR1 (100M) for 24h. The width of the scratch was measured using microscopy at 0 and 24h post-dosing, and the percentage of wound closure was calculated by comparing the scratch width at 24h to the initial scratch width.

NCM460 cells were treated with either DSS (20g/mL) or DSS (20g/mL)+NGR1 (100M) for 24h. Then, NCM460 cells were harvested and washed with PBS after experimental treatment. Cells were then suspended in a binding buffer containing Annexin V-fluorescein isothiocyanate (FITC) and propidium iodide (PI), and incubated in the dark at room temperature for 15min. Flow cytometry analysis was performed to detect apoptotic cells. The data were analyzed using Guava software, and the percentage of apoptotic cells was expressed.

Statistical analysis was performed using GraphPad Prism 9.0 software. Data were presented as meanstandard deviation (SD). Differences between groups were analyzed using one-way analysis of variance (ANOVA). P<0.05 was considered statistically significant. All experiments were repeated at least three times.

More here:
Notoginsenoside R1 promotes Lgr5+ stem cell and epithelium renovation in colitis mice via activating Wnt/-Catenin ... - Nature.com